US10022422B2 - Peptidomimetic macrocycles - Google Patents

Peptidomimetic macrocycles Download PDF

Info

Publication number
US10022422B2
US10022422B2 US15/093,373 US201615093373A US10022422B2 US 10022422 B2 US10022422 B2 US 10022422B2 US 201615093373 A US201615093373 A US 201615093373A US 10022422 B2 US10022422 B2 US 10022422B2
Authority
US
United States
Prior art keywords
virus
peptidomimetic
macrocycle
amino acid
methyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US15/093,373
Other versions
US20160250278A1 (en
Inventor
Huw M. Nash
David Allen Annis
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aileron Therapeutics Inc
Alleron Therapeutics Inc
Original Assignee
Alleron Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alleron Therapeutics Inc filed Critical Alleron Therapeutics Inc
Priority to US15/093,373 priority Critical patent/US10022422B2/en
Publication of US20160250278A1 publication Critical patent/US20160250278A1/en
Assigned to AILERON THERAPEUTICS, INC. reassignment AILERON THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NASH, HUW M., ANNIS, DAVID ALLEN
Priority to US16/002,977 priority patent/US20180339014A1/en
Application granted granted Critical
Publication of US10022422B2 publication Critical patent/US10022422B2/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/50Cyclic peptides containing at least one abnormal peptide link
    • C07K7/54Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/50Cyclic peptides containing at least one abnormal peptide link
    • C07K7/54Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring
    • C07K7/56Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring the cyclisation not occurring through 2,4-diamino-butanoic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • Seasonal influenza infection is a major health concern for first-world and developing countries alike. Each year in the United States, five- to twenty-percent of the population gets the flu, more than 200,000 people are hospitalized from flu complications, and about 36,000 people die from flu. Worldwide, influenza causes tens of millions of respiratory illnesses and 250,000 to 500,000 deaths each year. New strains of avian influenza that are transmissible to humans are a critical concern for global health because these flu strains could yield pandemic disease for which no immunity exists, potentially resulting in millions of fatalities. “Avian flu” refers to a pathogenic avian influenza subtype that is highly contagious among birds and causes high mortality among domestic poultry.
  • influenza viruses Since all influenza viruses have the ability to rapidly mutate, there is considerable concern that avian flu may be able to infect humans more easily and become communicable from one person to another. Also, avian flu virus strains have not infected many humans worldwide, so there is little or no immune protection against these strains in the human population; therefore, an influenza pandemic could easily occur if sustained avian flu virus transmission were to develop.
  • Influenza A viruses are divided into subtypes based on characteristics of two proteins, hemagglutinin (H) and neuraminidase (N), on the surface of the virus. There are 16 different hemagglutinin subtypes and 9 different neuraminidase subtypes, with H1N1 and H3N2 being the most common subtypes found in humans.
  • the avian flu virus refers to influenza A H5N1.
  • Influenza A is a negative-sense (3′ to 5′) single-stranded RNA virus.
  • RNA-dependent RNA polymerase RNA-dependent RNA polymerase to transcribe its genome to positive-sense RNA prior to translation.
  • RNA-dependent RNA polymerases have no mammalian counterpart, which renders species selectivity less problematic in the development of therapeutics that target this enzyme.
  • viral RNA-dependent RNA polymerases include polioviral 3Dpol, vesicular stomatitis virus L, and hepatitis C virus NS5b; the latter is an active target for development of hepatitis C antiviral therapies.
  • current flu targets e.g., neuraminidase for Tamiflu
  • influenza RNA polymerase is highly conserved and therefore less likely to suffer the resistance issues that current drugs face.
  • influenza protein RNA polymerase a critical enzyme for viral replication and a novel target for both therapeutic intervention and prophylaxis during influenza outbreaks.
  • the influenza RNA-dependent RNA polymerase is a heterotrimer of three subunits, PA, PB1, and PB2, with the 3 10 -helical N-terminal region of PB1 binding between the “jaws” of the PA protein.
  • the PB1 helix is thought to be important for complex formation and nuclear transport and inhibits influenza A viral replication by interfering with polymerase activity.
  • the PB2 subunit has also been shown to play an essential role in activity of the viral polymerase complex, for instance through contacts with the PB1 subunit. See Sugiyama et al, EMBO Journal, 2009, 28, 1803-1811. However, little is known about compounds capable of interfering with the binding and activity of these proteins. In general, there remains a need for therapeutic methods of treating viral diseases in which RNA-dependent RNA polymerases play a role, and for compositions and methods capable of modifying the activity such polymerases.
  • the present invention addresses these and other needs.
  • the present invention provides a peptidomimetic macrocycle capable of binding to a viral RNA-dependent RNA polymerase.
  • a macrocycle may, for example, be capable of disrupting the assembly of subunits of a viral RNA-dependent RNA polymerase complex.
  • such a macrocycle may compete with the binding of a peptide of the sequence MDVNPTLLFLKVPAQ (SEQ ID NO: 1) or MERIKELRNLM (SEQ ID NO: 2) to said viral RNA-dependent RNA polymerase.
  • a peptidomimetic macrocycle of the invention comprises an amino acid sequence which is at least about 60%, 80%, 90%, or 95% identical to the amino acid sequence MDVNPTLLFLKVPAQ (PB1) (SEQ ID NO: 1) or MERIKELRNLM (PB2) (SEQ ID NO: 2).
  • the amino acid sequence of said peptidomimetic macrocycle is identified and optimized for its ability to bind to either the PB1 peptide binding site of the PA protein or the PB2 peptide binding site of the PB1 protein, for example through affinity selection with the PA or PB1 target protein or by structure-based design, with such a mechanism of action being confirmed by biophysical/structural studies and/or competitive displacement assays with the PB1 or PB2 peptide.
  • the peptidomimetic macrocycle comprises a helix, such as a 3 10 helix or an ⁇ -helix.
  • the peptidomimetic macrocycle comprises an ⁇ , ⁇ -disubstituted amino acid.
  • a peptidomimetic macrocycle of the invention may comprise a crosslinker linking the ⁇ -positions of at least two amino acids. At least one of said two amino acids may be an ⁇ , ⁇ -disubstituted amino acid.
  • the peptidomimetic macrocycle has the formula:
  • each A, C, D, and E is independently a natural or non-natural amino acid
  • B is a natural or non-natural amino acid, amino acid analog
  • R 1 and R 2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 ;
  • L is a macrocycle-forming linker of the formula -L 1 -L 2 -;
  • L 1 and L 2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R 4 —K—R 4 —] n , each being optionally substituted with R 5 ;
  • each R 4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
  • each K is O, S, SO, SO 2 , CO, CO 2 , or CONR 3 ;
  • each R 5 is independently halogen, alkyl, —OR 6 , —N(R 6 ) 2 , —SR 6 , —SOR 6 , —SO 2 R 6 , —CO 2 R 6 , a fluorescent moiety, a radioisotope or a therapeutic agent;
  • each R 6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
  • R 7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 , or part of a cyclic structure with a D residue;
  • R 8 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 , or part of a cyclic structure with an E residue;
  • v and w are independently integers from 1-1000;
  • u, x, y and z are independently integers from 0-10;
  • n is an integer from 1-5.
  • the peptidomimetic macrocycle may comprise a crosslinker linking a backbone amino group of a first amino acid to a second amino acid within the peptidomimetic macrocycle.
  • the invention provides peptidomimetic macrocycles of the formula (IV) or (IVa):
  • each A, C, D, and E is independently a natural or non-natural amino acid
  • B is a natural or non-natural amino acid, amino acid analog
  • R 1 and R 2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-, or part of a cyclic structure with an E residue;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 ;
  • L 1 and L 2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R 4 —K—R 4 —] n , each being optionally substituted with R 5 ;
  • each R 4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
  • each K is O, S, SO, SO 2 , CO, CO 2 , or CONR 3 ;
  • each R 5 is independently halogen, alkyl, —OR 6 , —N(R 6 ) 2 , —SR 6 , —SOR 6 , —SO 2 R 6 , —CO 2 R 6 , a fluorescent moiety, a radioisotope or a therapeutic agent;
  • each R 6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
  • R 7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 ;
  • v and w are independently integers from 1-1000;
  • u, x, y and z are independently integers from 0-10;
  • n is an integer from 1-5.
  • x+y+z is 2, 3, 5 or 6.
  • the invention provides a method of treating influenza virus infection in a subject comprising administering to the subject a peptidomimetic macrocycle of the invention. Also provided is a method of preventing infection by an influenza virus in a subject comprising administering to the subject a peptidomimetic macrocycle of the invention, or a method of inhibiting the activity of the RNA-dependent RNA polymerase of an influenza virus in a subject comprising administering to the subject such a peptidomimetic macrocycle.
  • FIG. 1 a shows a bound PB1 helix in complex with the PA subunit of a RNA-dependent RNA polymerase.
  • Leu7 and Leu10 are candidate residues for i, i+3 macrocycle formation to stabilize a 3 10 helix.
  • FIG. 1 b shows a macrocycle derived from the sequence in FIG. 1 a.
  • FIG. 2 a shows the sequence of FIG. 1 a excised from its complex with the PA subunit or a RNA-dependent RNA polymerase.
  • FIG. 2 b shows the macrocycle derived from the sequence in FIG. 1 b excised from its complex with the PA subunit of a RNA-dependent RNA polymerase.
  • FIG. 3 describes the plasma stability of several peptidomimetic macrocycles of the invention.
  • FIG. 4 shows in vivo pharmacokinetic properties of several peptidomimetic macrocycles of the invention.
  • FIGS. 5 a -5 f illustrate in vivo pharmacokinetic properties of peptidomimetic macrocycles of the invention.
  • FIG. 6 shows selected pharmacokinetic parameters for several peptidomimetic macrocycles of the invention.
  • FIG. 7 illustrates pharmacokinetic properties for intravenous and subcutaneous modes of administration for a peptidomimetic macrocycle of the invention.
  • microcycle refers to a molecule having a chemical structure including a ring or cycle formed by at least 9 covalently bonded atoms.
  • peptidomimetic macrocycle or “crosslinked polypeptide” refers to a compound comprising a plurality of amino acid residues joined by a plurality of peptide bonds and at least one macrocycle-forming linker which forms a macrocycle between a first naturally-occurring or non-naturally-occurring amino acid residue (or analog) and a second naturally-occurring or non-naturally-occurring amino acid residue (or analog) within the same molecule.
  • Peptidomimetic macrocycle include embodiments where the macrocycle-forming linker connects the ⁇ carbon of the first amino acid residue (or analog) to the ⁇ carbon of the second amino acid residue (or analog).
  • the peptidomimetic macrocycles optionally include one or more non-peptide bonds between one or more amino acid residues and/or amino acid analog residues, and optionally include one or more non-naturally-occurring amino acid residues or amino acid analog residues in addition to any which form the macrocycle.
  • a “corresponding uncrosslinked polypeptide” when referred to in the context of a peptidomimetic macrocycle is understood to relate to a polypeptide of the same length as the macrocycle and comprising the equivalent natural amino acids of the wild-type sequence corresponding to the macrocycle.
  • the term “stability” refers to the maintenance of a defined secondary structure in solution by a peptidomimetic macrocycle of the invention as measured by circular dichroism, NMR or another biophysical measure, or resistance to proteolytic degradation in vitro or in vivo.
  • Non-limiting examples of secondary structures contemplated in this invention are helices, ⁇ -turns, and ⁇ -pleated sheets.
  • the term “helix” or “helical” is used to refer to any type of helical secondary structure, including 3 10 -helices, ⁇ -helices, and ⁇ -helices.
  • helical stability refers to the maintenance of helical structure by a peptidomimetic macrocycle of the invention as measured by circular dichroism or NMR.
  • the peptidomimetic macrocycles of the invention exhibit at least a 1.25, 1.5, 1.75 or 2-fold increase in helicity as determined by circular dichroism compared to a corresponding uncrosslinked macrocycle.
  • amino acid refers to a molecule containing both an amino group and a carboxyl group bound to a carbon which is designated the ⁇ -carbon.
  • Suitable amino acids include, without limitation, both the D- and L-isomers of the naturally-occurring amino acids, as well as non-naturally occurring amino acids prepared by organic synthesis or other metabolic routes. Unless the context specifically indicates otherwise, the term amino acid, as used herein, is intended to include amino acid analogs.
  • naturally occurring amino acid refers to any one of the twenty amino acids commonly found in peptides synthesized in nature, and known by the one letter abbreviations A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y and V.
  • amino acid analog or “non-natural amino acid” refers to a molecule which is structurally similar to an amino acid and which can be substituted for an amino acid in the formation of a peptidomimetic macrocycle.
  • Amino acid analogs include, without limitation, compounds which are structurally identical to an amino acid, as defined herein, except for the inclusion of one or more additional methylene groups between the amino and carboxyl group (e.g., ⁇ -amino ⁇ -carboxy acids), or for the substitution of the amino or carboxy group by a similarly reactive group (e.g., substitution of the primary amine with a secondary or tertiary amine, or substitution or the carboxy group with an ester).
  • non-essential amino acid residue is a residue that can be altered from the wild-type sequence of a polypeptide (e.g., a BH3 domain or the p53 MDM2 binding domain) without abolishing or substantially altering its essential biological or biochemical activity (e.g., receptor binding or activation).
  • essential amino acid residue is a residue that, when altered from the wild-type sequence of the polypeptide, results in abolishing or substantially abolishing the polypeptide's essential biological or biochemical activity.
  • a “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., K, R, H), acidic side chains (e.g., D, E), uncharged polar side chains (e.g., G, N, Q, S, T, Y, C), nonpolar side chains (e.g., A, V, L, I, P, F, M, W), beta-branched side chains (e.g., T, V, I) and aromatic side chains (e.g., Y, F, W, H).
  • basic side chains e.g., K, R, H
  • acidic side chains e.g., D, E
  • uncharged polar side chains e.g., G, N, Q, S, T, Y, C
  • nonpolar side chains e.g., A, V, L
  • a predicted nonessential amino acid residue in a polypeptide is preferably replaced with another amino acid residue from the same side chain family.
  • Other examples of acceptable substitutions are substitutions based on isosteric considerations (e.g. norleucine for methionine) or other properties (e.g. 2-thienylalanine for phenylalanine).
  • member refers to the atoms that form or can form the macrocycle, and excludes substituent or side chain atoms.
  • cyclodecane, 1,2-difluoro-decane and 1,3-dimethyl cyclodecane are all considered ten-membered macrocycles as the hydrogen or fluoro substituents or methyl side chains do not participate in forming the macrocycle.
  • amino acid side chain refers to a moiety attached to the ⁇ -carbon in an amino acid.
  • amino acid side chain for alanine is methyl
  • amino acid side chain for phenylalanine is phenylmethyl
  • amino acid side chain for cysteine is thiomethyl
  • amino acid side chain for aspartate is carboxymethyl
  • amino acid side chain for tyrosine is 4-hydroxyphenylmethyl
  • Other non-naturally occurring amino acid side chains are also included, for example, those that occur in nature (e.g., an amino acid metabolite) or those that are made synthetically (e.g., an ⁇ , ⁇ di-substituted amino acid).
  • ⁇ , ⁇ di-substituted amino acid refers to a molecule or moiety containing both an amino group and a carboxyl group bound to a carbon (the ⁇ -carbon) that is attached to two natural or non-natural amino acid side chains.
  • polypeptide encompasses two or more naturally or non-naturally-occurring amino acids joined by a covalent bond (e.g., an amide bond).
  • Polypeptides as described herein include full length proteins (e.g., fully processed proteins) as well as shorter amino acid sequences (e.g., fragments of naturally-occurring proteins or synthetic polypeptide fragments).
  • microcyclization reagent or “macrocycle-forming reagent” as used herein refers to any reagent which may be used to prepare a peptidomimetic macrocycle of the invention by mediating the reaction between two reactive groups.
  • Reactive groups may be, for example, an azide and alkyne
  • macrocyclization reagents include, without limitation, Cu reagents such as reagents which provide a reactive Cu(I) species, such as CuBr, CuI or CuOTf, as well as Cu(II) salts such as Cu(CO 2 CH 3 ) 2 , CuSO 4 , and CuCl 2 that can be converted in situ to an active Cu(I) reagent by the addition of a reducing agent such as ascorbic acid or sodium ascorbate.
  • Macrocyclization reagents may additionally include, for example, Ru reagents known in the art such as Cp*RuCl(PPh 3 ) 2 , [Cp*RuCl] 4 or other Ru reagents which may provide a reactive Ru(II) species.
  • the reactive groups are terminal olefins.
  • the macrocyclization reagents or macrocycle-forming reagents are metathesis catalysts including, but not limited to, stabilized, late transition metal carbene complex catalysts such as Group VIII transition metal carbene catalysts.
  • such catalysts are Ru and Os metal centers having a +2 oxidation state, an electron count of 16 and pentacoordinated.
  • the reactive groups are thiol groups.
  • the macrocyclization reagent is, for example, a linker functionalized with two thiol-reactive groups such as halogen groups.
  • halo or halogen refers to fluorine, chlorine, bromine or iodine or a radical thereof.
  • alkyl refers to a hydrocarbon chain that is a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C 1 -C 10 indicates that the group has from 1 to 10 (inclusive) carbon atoms in it. In the absence of any numerical designation, “alkyl” is a chain (straight or branched) having 1 to 20 (inclusive) carbon atoms in it.
  • alkylene refers to a divalent alkyl (i.e., —R—).
  • alkenyl refers to a hydrocarbon chain that is a straight chain or branched chain having one or more carbon-carbon double bonds.
  • the alkenyl moiety contains the indicated number of carbon atoms. For example, C 2 -C 10 indicates that the group has from 2 to 10 (inclusive) carbon atoms in it.
  • lower alkenyl refers to a C 2 -C 6 alkenyl chain. In the absence of any numerical designation, “alkenyl” is a chain (straight or branched) having 2 to 20 (inclusive) carbon atoms in it.
  • alkynyl refers to a hydrocarbon chain that is a straight chain or branched chain having one or more carbon-carbon triple bonds.
  • the alkynyl moiety contains the indicated number of carbon atoms.
  • C 2 -C 10 indicates that the group has from 2 to 10 (inclusive) carbon atoms in it.
  • lower alkynyl refers to a C 2 -C 6 alkynyl chain.
  • alkynyl is a chain (straight or branched) having 2 to 20 (inclusive) carbon atoms in it.
  • aryl refers to a 6-carbon monocyclic or 10-carbon bicyclic aromatic ring system wherein 0, 1, 2, 3, or 4 atoms of each ring are substituted by a substituent.
  • aryl groups include phenyl, naphthyl and the like.
  • arylalkyl or the term “aralkyl” refers to alkyl substituted with an aryl.
  • arylalkoxy refers to an alkoxy substituted with aryl.
  • Arylalkyl refers to an aryl group, as defined above, wherein one of the aryl group's hydrogen atoms has been replaced with a C 1 -C 5 alkyl group, as defined above.
  • Representative examples of an arylalkyl group include, but are not limited to, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 2-propylphenyl, 3-propylphenyl, 4-propylphenyl, 2-butylphenyl, 3-butylphenyl, 4-butylphenyl, 2-pentylphenyl, 3-pentylphenyl, 4-pentylphenyl, 2-isopropylphenyl, 3-isopropylphenyl, 4-isopropylphenyl, 2-isobutylphenyl, 3-isobutylphenyl, 4-isopropylphenyl
  • Arylamido refers to an aryl group, as defined above, wherein one of the aryl group's hydrogen atoms has been replaced with one or more —C(O)NH 2 groups.
  • Representative examples of an arylamido group include 2-C(O)NH2-phenyl, 3-C(O)NH 2 -phenyl, 4-C(O)NH 2 -phenyl, 2-C(O)NH 2 -pyridyl, 3-C(O)NH 2 -pyridyl, and 4-C(O)NH 2 -pyridyl,
  • Alkylheterocycle refers to a C 1 -C 5 alkyl group, as defined above, wherein one of the C 1 -C 5 alkyl group's hydrogen atoms has been replaced with a heterocycle.
  • Representative examples of an alkylheterocycle group include, but are not limited to, —CH 2 CH 2 -morpholine, —CH 2 CH 2 -piperidine, —CH 2 CH 2 CH 2 -morpholine, and —CH 2 CH 2 CH 2 -imidazole.
  • Alkylamido refers to a C 1 -C 5 alkyl group, as defined above, wherein one of the C 1 -C 5 alkyl group's hydrogen atoms has been replaced with a —C(O)NH 2 group.
  • an alkylamido group include, but are not limited to, —CH 2 —C(O)NH 2 , —CH 2 CH 2 —C(O)NH 2 , —CH 2 CH 2 CH 2 C(O)NH 2 , —CH 2 CH 2 CH 2 CH 2 C(O)NH 2 , —CH 2 CH 2 CH 2 CH 2 C(O)NH 2 , —CH 2 CH(C(O)NH 2 )CH 3 , —CH 2 CH(C(O)NH 2 )CH 2 CH 3 , —CH(C(O)NH 2 )CH 2 CH 3 , —C(CH 3 ) 2 CH 2 C(O)NH 2 , —CH 2 —CH 2 —NH—C(O)—CH 3 , —CH 2 —CH 2 —NH—C(O)—CH 3 , —CH 2 —CH 2 —NH—C(O)—CH 3 , —CH 2 —CH 2 —NH—C(O)—CH 3
  • Alkanol refers to a C 1 -C 5 alkyl group, as defined above, wherein one of the C 1 -C 5 alkyl group's hydrogen atoms has been replaced with a hydroxyl group.
  • Representative examples of an alkanol group include, but are not limited to, —CH 2 OH, —CH 2 CH 2 OH, —CH 2 CH 2 CH 2 OH, —CH 2 CH 2 CH 2 CH 2 OH, —CH 2 CH 2 CH 2 CH 2 CH 2 OH, —CH 2 CH(OH)CH 3 , —CH 2 CH(OH)CH 2 CH 3 , —CH(OH)CH 3 and —C(CH 3 ) 2 CH 2 OH.
  • Alkylcarboxy refers to a C 1 -C 5 alkyl group, as defined above, wherein one of the C 1 -C 5 alkyl group's hydrogen atoms has been replaced with a —COOH group.
  • Representative examples of an alkylcarboxy group include, but are not limited to, —CH 2 COOH, —CH 2 CH 2 COOH, —CH 2 CH 2 CH 2 COOH, —CH 2 CH 2 CH 2 CH 2 COOH, —CH 2 CH(COOH)CH 3 , —CH 2 CH 2 CH 2 CH 2 COOH, —CH 2 CH(COOH)CH 2 CH 3 , —CH(COOH)CH 2 CH 3 and —C(CH 3 ) 2 CH 2 COOH.
  • cycloalkyl as employed herein includes saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12 carbons, preferably 3 to 8 carbons, and more preferably 3 to 6 carbons, wherein the cycloalkyl group additionally is optionally substituted.
  • Some cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl.
  • heteroaryl refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of O, N, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2, 3, or 4 atoms of each ring are substituted by a substituent.
  • heteroaryl groups include pyridyl, furyl or furanyl, imidazolyl, benzimidazolyl, pyrimidinyl, thiophenyl or thienyl, quinolinyl, indolyl, thiazolyl, and the like.
  • heteroarylalkyl or the term “heteroaralkyl” refers to an alkyl substituted with a heteroaryl.
  • heteroarylalkoxy refers to an alkoxy substituted with heteroaryl.
  • heteroarylalkyl or the term “heteroaralkyl” refers to an alkyl substituted with a heteroaryl.
  • heteroarylalkoxy refers to an alkoxy substituted with heteroaryl.
  • heterocyclyl refers to a nonaromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of O, N, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring are substituted by a substituent.
  • heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like.
  • substituted refers to a group replacing a second atom or group such as a hydrogen atom on any molecule, compound or moiety.
  • Suitable substituents include, without limitation, halo, hydroxy, mercapto, oxo, nitro, haloalkyl, alkyl, alkaryl, aryl, aralkyl, alkoxy, thioalkoxy, aryloxy, amino, alkoxycarbonyl, amido, carboxy, alkanesulfonyl, alkylcarbonyl, and cyano groups.
  • the compounds of this invention contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of these compounds are included in the present invention unless expressly provided otherwise.
  • the compounds of this invention are also represented in multiple tautomeric forms, in such instances, the invention includes all tautomeric forms of the compounds described herein (e.g., if alkylation of a ring system results in alkylation at multiple sites, the invention includes all such reaction products). All such isomeric forms of such compounds are included in the present invention unless expressly provided otherwise. All crystal forms of the compounds described herein are included in the present invention unless expressly provided otherwise.
  • the terms “increase” and “decrease” mean, respectively, to cause a statistically significantly (i.e., p ⁇ 0.1) increase or decrease of at least 5%.
  • variable is equal to any of the values within that range.
  • variable is equal to any integer value within the numerical range, including the end-points of the range.
  • variable is equal to any real value within the numerical range, including the end-points of the range.
  • a variable which is described as having values between 0 and 2 takes the values 0, 1 or 2 if the variable is inherently discrete, and takes the values 0.0, 0.1, 0.01, 0.001, or any other real values ⁇ 0 and ⁇ 2 if the variable is inherently continuous.
  • on average represents the mean value derived from performing at least three independent replicates for each data point.
  • biological activity encompasses structural and functional properties of a macrocycle of the invention.
  • Biological activity is, for example, structural stability, helicity (including, e.g. alpha-helicity), affinity for a target, resistance to proteolytic degradation, cell penetrability, intracellular stability, in vivo stability, or any combination thereof.
  • peptidomimetic macrocycles are prepared that target or interact with proteins that a virus needs for infection or replication within a host cell.
  • viruses may be, for example, influenza viruses belonging to Orthomyxoviridae family of viruses. This family also includes Thogoto viruses and Dhoriviruses.
  • Influenza type A viruses infect people, birds, pigs, horses, seals and other animals, but wild birds are the natural hosts for these viruses.
  • Influenza type A viruses are divided into subtypes and named on the basis of two proteins on the surface of the virus: hemagglutinin (HA) and neuraminidase (NA).
  • HA hemagglutinin
  • NA neuraminidase
  • an “H7N2 virus” designates an influenza A subtype that has an HA 7 protein and an NA 2 protein.
  • an “H5N1” virus has an HA 5 protein and an NA 1 protein.
  • Only some influenza A subtypes i.e., H1N1, H1N2, and H3N2 are currently in general circulation among people. Other subtypes are found most commonly in other animal species.
  • H7N7 and H3N8 viruses cause illness in horses, and H3N8 also has recently been shown to cause illness in dogs.
  • Antiviral agents according to the invention can be used to protect high-risk groups (hospital units, institutes caring for elderly, immuno-suppressed individuals), and on a case by case basis.
  • a potential use for antiviral agents is to limit the spread and severity of the future pandemics whether caused by avian H5N1 or other strains of influenza virus.
  • Avian influenza A viruses of the subtypes H5 and H7, including H5N1, H7N7, and H7N3 viruses have been associated with high pathogenicity, and human infection with these viruses have ranged from mild (H7N3, H7N7) to severe and fatal disease (H7N7, H5N1).
  • Influenza B viruses are usually found in humans but can also infect seals. Unlike influenza A viruses, these viruses are not classified according to subtype. Influenza B viruses can cause morbidity and mortality among humans, but in general are associated with less severe epidemics than influenza A viruses. Although influenza type B viruses can cause human epidemics, they have not caused pandemics.
  • Influenza type C viruses cause mild illness in humans and do not cause epidemics or pandemics. These viruses can also infect dogs and pigs. These viruses are not classified according to subtype.
  • Influenza viruses differ from each other in respect to cell surface receptor specificity and cell tropism, however they use common entry pathways. Charting these pathways and identification of host cell proteins involved in virus influenza transmission, entry, replication, biosynthesis, assembly, or exit allows the development of general agents against existing and emerging strains of influenza.
  • the agents may also prove useful against unrelated viruses that use similar pathways. For example, the agents may protect airway epithelial cells against a number of different viruses in addition to influenza viruses.
  • the targeted virus is an adenovirus.
  • Adenoviruses most commonly cause respiratory illness; symptoms of respiratory illness caused by adenovirus infection range from the common cold syndrome to pneumonia, croup, and bronchitis. Patients with compromised immune systems are especially susceptible to severe complications of adenovirus infection.
  • Acute respiratory disease (ARD), first recognized among military recruits during World War II, can be caused by adenovirus infections during conditions of crowding and stress.
  • Adenoviruses are medium-sized (90-100 nm), nonenveloped icosohedral viruses containing double-stranded DNA.
  • immunologically distinct types (6 subgenera: A through F) that can cause human infections.
  • Adenoviruses are unusually stable to chemical or physical agents and adverse pH conditions, allowing for prolonged survival outside of the body. Some adenoviruses, such as AD2 and Ad5 (species C) use clathrin mediated endocytosis and macropinocytosis for infectious entry. Other adenoviruses, such as Ad3 (species B) use dynamin dependent endocytosis and macropinocytosis for infectious entry.
  • the targeted virus is a respiratory syncytial virus (RSV).
  • RSV respiratory syncytial virus
  • Illness begins most frequently with fever, runny nose, cough, and sometimes wheezing.
  • RSV infection between 25% and 40% of infants and young children have signs or symptoms of bronchiolitis or pneumonia, and 0.5% to 2% require hospitalization.
  • Most children recover from illness in 8 to 15 days. The majority of children hospitalized for RSV infection are under 6 months of age.
  • RSV also causes repeated infections throughout life, usually associated with moderate-to-severe cold-like symptoms; however, severe lower respiratory tract disease may occur at any age, especially among the elderly or among those with compromised cardiac, pulmonary, or immune systems.
  • RSV is a negative-sense, enveloped RNA virus.
  • the virion is variable in shape and size (average diameter of between 120 and 300 nm), is unstable in the environment (surviving only a few hours on environmental surfaces), and is readily inactivated with soap and water and disinfectants.
  • the targeted virus is a human parainfluenza virus (HPIV).
  • HPIVs arc second to respiratory syncytial virus (RSV) as a common cause of lower respiratory tract disease in young children. Similar to RSV, HPIVs can cause repeated infections throughout life, usually manifested by an upper respiratory tract illness (e.g., a cold and/or sore throat). HPIVs can also cause serious lower respiratory tract disease with repeat infection (e.g., pneumonia, bronchitis, and bronchiolitis), especially among the elderly, and among patients with compromised immune systems. Each of the four HPIVs has different clinical and epidemiologic features.
  • HPIV-1 and HPIV-2 are distinctive clinical feature of HPIV-1 and HPIV-2.
  • croup i.e., laryngotracheobronchitis
  • HPIV-1 is the leading cause of croup in children, whereas HPIV-2 is less frequently detected.
  • HPIV-1 and -2 can cause other upper and lower respiratory tract illnesses.
  • HPIV-3 is more often associated with bronchiolitis and pneumonia.
  • HPIV-4 is infrequently detected, possibly because it is less likely to cause severe disease.
  • the incubation period for HPIVs is generally from 1 to 7 days.
  • HPIVs are negative-sense, single-stranded RNA viruses that possess fusion and hemagglutinin-neuraminidase glycoprotein “spikes” on their surface.
  • HPIV HPIV
  • subtypes 4a and 4h
  • the virion varies in size (average diameter between 150 and 300 nm) and shape, is unstable in the environment (surviving a few hours on environmental surfaces), and is readily inactivated with soap and water.
  • the targeted virus is a coronavirus.
  • Coronavirus is a genus of animal virus belonging to the family Coronaviridae. Coronaviruses are enveloped viruses with a positive-sense single-stranded RNA genome and a helical symmetry. The genomic size of coronaviruses ranges from approximately 16 to 31 kilobases, extraordinarily large for an RNA virus.
  • the name “coronavirus” is derived from the Latin corona, meaning crown, as the virus envelope appears under electron microscopy to be crowned by a characteristic ring of small bulbous structures. This morphology is actually formed by the viral spike peplomers, which are proteins that populate the surface of the virus and determine host tropism.
  • Coronaviruses are grouped in the order Nidovirales, named for the Latin nidus, meaning nest, as all viruses in this order produce a 3′ co-terminal nested set of subgenomic mRNA's during infection. Proteins that contribute to the overall structure of all coronaviruses are the spike, envelope, membrane and nucleocapsid. In the specific case of SARS a defined receptor-binding domain on S mediates the attachment of the virus to its cellular receptor, angiotensin-converting enzyme 2.
  • the targeted virus is a rhinovirus.
  • Rhinovirus (from the Greek rhin-, which means “nose”) is a genus of the Picornaviridae family of viruses. Rhinoviruses are the most common viral infective agents in humans, and a causative agent of the common cold. There are over 105 serologic virus types that cause cold symptoms, and rhinoviruses are responsible for approximately 50% of all cases. Rhinoviruses have single-stranded positive sense RNA genomes of between 7.2 and 8.5 kb in length. At the 5′ end of the genome is a virus-encoded protein, and like mammalian mRNA, there is a 3′ poly-A tail. Structural proteins are encoded in the 5′ region of the genome and non structural at the end. This is the same for all picornaviruses. The viral particles themselves are not enveloped and are icosahedral in structure.
  • Any secondary structure of a viral protein can form the basis of the methods of the invention.
  • a viral proteins comprising a secondary structure which is a helix may be used to design peptidomimetic macrocycles based on said helix.
  • the peptidomimetic macrocycle of the invention is designed based on the PB1 or PB2 sequence of an influenza virus.
  • the PB1 sequence is highly conserved across all known strains of influenza A virus, which may result in less drug resistance should than that observed with the current standard of care.
  • An alignment of the first 25 N-terminal amino acids of PB1 from the NCBI data bank's 2,485 influenza A virus strains (Ghanem, 2007) demonstrates the remarkable sequence conservation in the PA interaction domain of PB1. Therefore, antiviral therapies based on the PB1 sequence may block most if not all, influenza A virus strains. Additionally, sequence modification of a peptidomimetic macrocycle based on these few variations in PB1 may enable an antiviral cocktail of PB1 inhibitors to eliminate resistance due to escape mutants.
  • a peptidomimetic macrocycle of the invention has the Formula (I):
  • each A, C, D, and E is independently a natural or non-natural amino acid
  • B is a natural or non-natural amino acid, amino acid analog
  • R 1 and R 2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 ;
  • L is a macrocycle-forming linker of the formula -L 1 -L 2 -;
  • L 1 and L 2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R 4 —K—R 4 —] n , each being optionally substituted with R 5 ;
  • each R 4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
  • each K is O, S, SO, SO 2 , CO, CO 2 , or CONR 3 ;
  • each R 5 is independently halogen, alkyl, —OR 6 , —N(R 6 ) 2 , —SR 6 , —SOR 6 , —SO 2 R 6 , —CO 2 R 6 , a fluorescent moiety, a radioisotope or a therapeutic agent;
  • each R 6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
  • R 7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 , or part of a cyclic structure with a D residue;
  • R 8 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 , or part of a cyclic structure with an E residue;
  • v and w are independently integers from 1-1000;
  • u, x, y and z are independently integers from 0-10;
  • n is an integer from 1-5.
  • At least one of R 1 and R 2 is alkyl, unsubstituted or substituted with halo-. In another example, both R 1 and R 2 are independently alkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of R 4 and R 2 is methyl. In other embodiments, R 1 and R 2 are methyl.
  • x+y+z is at least 2. In other embodiments of the invention, x+y+z is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • Each occurrence of A, B, C, D or E in a macrocycle or macrocycle precursor of the invention is independently selected.
  • a sequence represented by the formula [A] x when x is 3, encompasses embodiments where the amino acids are not identical, e.g. Gln-Asp-Ala as well as embodiments where the amino acids are identical, e.g. Gln-Gln-Gln. This applies for any value of x, y, or z in the indicated ranges.
  • each compound of the invention may encompass peptidomimetic macrocycles which are the same or different.
  • a compound of the invention may comprise peptidomimetic macrocycles comprising different linker lengths or chemical compositions.
  • the peptidomimetic macrocycle of the invention comprises a secondary structure which is a helix and R 8 is —H, allowing intrahelical hydrogen bonding.
  • at least one of A, B, C, D or E is an ⁇ , ⁇ -disubstituted amino acid.
  • B is an ⁇ , ⁇ -disubstituted amino acid.
  • at least one of A, B, C, D or E is 2-aminoisobutyric acid.
  • at least one of A, B, C, D or E is
  • the length of the macrocycle-forming linker L as measured from a first C ⁇ to a second C ⁇ is selected to stabilize a desired secondary peptide structure, such as a helix formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first C ⁇ to a second C ⁇ .
  • the peptidomimetic macrocycle of Formula (I) is:
  • each R 1 and R 2 is independently H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo.
  • the peptidomimetic macrocycle of Formula (I) is:
  • the peptidomimetic macrocycles of the invention have the Formula (II):
  • each A, C, D, and E is independently a natural or non-natural amino acid
  • B is a natural or non-natural amino acid, amino acid analog
  • R 1 and R 2 are independently H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 ;
  • L is a macrocycle-forming linker of the formula
  • L 1 , L 2 and L 3 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R 4 —K—R 4 —] n , each being optionally substituted with R 5 ;
  • each R 4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
  • each K is O, S, SO, SO 2 , CO, CO 2 , or CONR 3 ;
  • each R 5 is independently halogen, alkyl, —OR 6 , —N(R 6 ) 2 , —SR 6 , —SOR 6 , —SO 2 R 6 , —CO 2 R 6 , a fluorescent moiety, a radioisotope or a therapeutic agent;
  • each R 6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
  • R 7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 , or part of a cyclic structure with a D residue;
  • R 8 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 , or part of a cyclic structure with an F residue;
  • v and w are independently integers from 1-1000;
  • u, x, y and z are independently integers from 0-10;
  • n is an integer from 1-5.
  • At least one of R 1 is alkyl, unsubstituted or substituted with halo-. In another example, both R 1 and R 2 are independently alkyl, unsubstituted or substituted with halo. In some embodiments, at least one of R 1 and R 2 is methyl. In other embodiments, R 1 and R 2 are methyl.
  • x+y+z is at least 2. In other embodiments of the invention, x+y+z is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • Each occurrence of A, B, C, D or E in a macrocycle or macrocycle precursor of the invention is independently selected.
  • a sequence represented by the formula [A] x when x is 3, encompasses embodiments where the amino acids are not identical, e.g. Gln Asp Ala as well as embodiments where the amino acids are identical, e.g. Gln Gln Gln. This applies for any value of x, y, or z in the indicated ranges.
  • the peptidomimetic macrocycle of the invention comprises a secondary structure which is a helix and R 8 is —H, allowing intrahelical hydrogen bonding.
  • at least one of A, B, C, D or F is an ⁇ , ⁇ -disubstituted amino acid.
  • B is an ⁇ , ⁇ -disubstituted amino acid.
  • at least one of A, B, C, D or E is 2-aminoisobutyric acid.
  • at least one of A, B, C, D or E is
  • the length of the macrocycle-forming linker L as measured from a first C ⁇ to a second C ⁇ is selected to stabilize a desired secondary peptide structure, such as a helix formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first C ⁇ to a second C ⁇ .
  • the invention provides peptidomimetic macrocycles of Formula (III):
  • each A, C, D, and E is independently a natural or non-natural amino acid
  • B is a natural or non-natural amino acid, amino acid analog
  • R 1 and R 2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, unsubstituted or substituted with R 5 ;
  • L 1 , L 2 , L 3 and L 4 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene or [—R 4 —K—R 4 —]n, each being unsubstituted or substituted with R 5 ;
  • K is O, S, SO, SO 2 , CO, CO 2 , or CONR 3 ;
  • each R 4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
  • each R 5 is independently halogen, alkyl, —OR 6 , —N(R 6 ) 2 , —SR 6 , —SOR 6 , —SO 2 R 6 , —CO 2 R 6 , a fluorescent moiety, a radioisotope or a therapeutic agent;
  • each R 6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
  • R 7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, unsubstituted or substituted with R 5 , or part of a cyclic structure with a D residue;
  • R 8 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, unsubstituted or substituted with R 5 , or part of a cyclic structure with an E residue;
  • v and w are independently integers from 1-1000;
  • u, x, y and z are independently integers from 0-10;
  • n is an integer from 1-5.
  • At least one of R 1 and R 2 is alkyl, unsubstituted or substituted with halo-. In another example, both R 1 and R 2 are independently alkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of R 1 and R 2 is methyl. In other embodiments, R 1 and R 2 are methyl.
  • x+y+z is at least 2. In other embodiments of the invention, x+y+z is 3, 4, 5, 6, 7, 8, 9 or 10.
  • Each occurrence of A, B, C, D or E in a macrocycle or macrocycle precursor of the invention is independently selected.
  • a sequence represented by the formula [A] x when x is 3, encompasses embodiments where the amino acids are not identical, e.g. Gln-Asp-Ala as well as embodiments where the amino acids are identical, e.g. Gln-Gln-Gln. This applies for any value of x, y, or z in the indicated ranges.
  • the peptidomimetic macrocycle of the invention comprises a secondary structure which is a helix and R 8 is —H, allowing intrahelical hydrogen bonding.
  • at least one of A, B, C, D or E is an ⁇ , ⁇ -disubstituted amino acid.
  • B is an ⁇ , ⁇ -disubstituted amino acid.
  • at least one of A, B, C, D or E is 2-aminoisobutyric acid.
  • at least one of A, B, C, D or E is
  • the length of the macrocycle-forming linker [-L 1 -S-L 2 -S-L 3 -] as measured from a first C ⁇ to a second C ⁇ is selected to stabilize a desired secondary peptide structure, such as a helix (including, but not limited to a 3 10 helix or an ⁇ -helix) formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first C ⁇ to a second C ⁇ .
  • a desired secondary peptide structure such as a helix (including, but not limited to a 3 10 helix or an ⁇ -helix) formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first C ⁇ to a second C ⁇ .
  • Macrocycles or macrocycle precursors are synthesized, for example, by solution phase or solid-phase methods, and can contain both naturally-occurring and non-naturally-occurring amino acids. See, for example, Hunt, “The Non-Protein Amino Acids” in Chemistry and Biochemistry of the Amino Acids , edited by G. C. Barrett, Chapman and Hall, 1985.
  • the thiol moieties are the side chains of the amino acid residues L-cysteine, D-cysteine, ⁇ -methyl-L cysteine, ⁇ -methyl-D-cysteine, L-homocysteine, D-homocysteine, ⁇ -methyl-L-homocysteine or ⁇ -methyl-D-homocysteine.
  • a bis-alkylating reagent is of the general formula X-L 2 -Y wherein L 2 is a linker moiety and X and Y are leaving groups that are displaced by —SH moieties to form bonds with L 2 .
  • X and Y are halogens such as I, Br, or Cl.
  • D and/or E in the compound of Formula I, II or III are further modified in order to facilitate cellular uptake.
  • lipidating or PEGylating a peptidomimetic macrocycle facilitates cellular uptake, increases bioavailability, increases blood circulation, alters pharmacokinetics, decreases immunogenicity and/or decreases the needed frequency of administration.
  • At least one of [D] and [E] in the compound of Formula I, II or III represents a moiety comprising an additional macrocycle-forming linker such that the peptidomimetic macrocycle comprises at least two macrocycle-forming linkers.
  • a peptidomimetic macrocycle comprises two macrocycle-forming linkers.
  • any of the macrocycle-forming linkers described herein may be used in any combination with any of the sequences shown in Tables 1-4 and also with any of the R substituents indicated herein.
  • the peptidomimetic macrocycle of the invention comprises at least one helical motif, such as a 3 10 or an ⁇ -helix motif.
  • A, B and/or C in the compound of Formula I, II or III include one or more helices.
  • helices include between 3 and 4 amino acid residues per turn.
  • the helix of the peptidomimetic macrocycle includes 1 to 5 turns and, therefore, 3 to 20 amino acid residues.
  • the helix includes 1 turn, 2 turns, 3 turns, 4 turns, or 5 turns.
  • the macrocycle-forming linker stabilizes a helix motif included within the peptidomimetic macrocycle.
  • the length of the macrocycle-forming linker L from a first C ⁇ to a second C ⁇ is selected to increase the stability of a helix.
  • the macrocycle-forming linker spans from 1 turn to 5 turns of the helix. In some embodiments, the macrocycle-forming linker spans approximately 1 turn, 2 turns, 3 turns, 4 turns, or 5 turns of the helix. In some embodiments, the length of the macrocycle-forming linker is approximately 5 ⁇ to 9 ⁇ per turn of the helix, or approximately 6 ⁇ to 8 ⁇ per turn of the helix.
  • the length is equal to approximately 5 carbon-carbon bonds to 13 carbon-carbon bonds, approximately 7 carbon-carbon bonds to 11 carbon-carbon bonds, or approximately 9 carbon-carbon bonds.
  • the length is equal to approximately 8 carbon-carbon bonds to 16 carbon-carbon bonds, approximately 10 carbon-carbon bonds to 14 carbon-carbon bonds, or approximately 12 carbon-carbon bonds.
  • the macrocycle-forming linker spans approximately 3 turns of a helix, the length is equal to approximately 14 carbon-carbon bonds to 22 carbon-carbon bonds, approximately 16 carbon-carbon bonds to 20 carbon-carbon bonds, or approximately 18 carbon-carbon bonds.
  • the length is equal to approximately 20 carbon-carbon bonds to 28 carbon-carbon bonds, approximately 22 carbon-carbon bonds to 26 carbon-carbon bonds, or approximately 24 carbon-carbon bonds.
  • the macrocycle-forming linker spans approximately 5 turns of a helix, the length is equal to approximately 26 carbon-carbon bonds to 34 carbon-carbon bonds, approximately 28 carbon-carbon bonds to 32 carbon-carbon bonds, or approximately 30 carbon-carbon bonds.
  • the linkage contains approximately 4 atoms to 12 atoms, approximately 6 atoms to 10 atoms, or approximately 8 atoms.
  • the linkage contains approximately 7 atoms to 15 atoms, approximately 9 atoms to 13 atoms, or approximately 11 atoms.
  • the linkage contains approximately 13 atoms to 21 atoms, approximately 15 atoms to 19 atoms, or approximately 17 atoms.
  • the linkage contains approximately 19 atoms to 27 atoms, approximately 21 atoms to 25 atoms, or approximately 23 atoms.
  • the linkage contains approximately 25 atoms to 33 atoms, approximately 27 atoms to 31 atoms, or approximately 29 atoms.
  • the resulting macrocycle forms a ring containing approximately 17 members to 25 members, approximately 19 members to 23 members, or approximately 21 members.
  • the macrocycle-forming linker spans approximately 2 turns of the helix, the resulting macrocycle forms a ring containing approximately 29 members to 37 members, approximately 31 members to 35 members, or approximately 33 members.
  • the resulting macrocycle forms a ring containing approximately 44 members to 52 members, approximately 46 members to 50 members, or approximately 48 members.
  • the resulting macrocycle forms a ring containing approximately 59 members to 67 members, approximately 61 members to 65 members, or approximately 63 members.
  • the macrocycle-forming linker spans approximately 5 turns of the helix, the resulting macrocycle forms a ring containing approximately 74 members to 82 members, approximately 76 members to 80 members, or approximately 78 members.
  • the invention provides peptidomimetic macrocycles of Formula (IV) or (IVa):
  • each A, C, D, and E is independently a natural or non-natural amino acid
  • B is a natural or non-natural amino acid, amino acid analog
  • R 1 and R 2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-, or part of a cyclic structure with an E residue;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 ;
  • L is a macrocycle-forming linker of the formula -L 4 -L 2 -;
  • L 1 and L 2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R 4 —K—R 4 —] n , each being optionally substituted with R 5 ;
  • each R 4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
  • each K is O, S, SO, SO 2 , CO, CO 2 , or CONR 3 ;
  • each R 5 is independently halogen, alkyl, —OR 6 , —N(R 6 ) 2 , —SR 6 , —SOR 6 , —SO 2 R 6 , —CO 2 R 6 , a fluorescent moiety, a radioisotope or a therapeutic agent;
  • each R 6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
  • R 7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R 5 ;
  • v and w are independently integers from 1-1000;
  • u, x, y and z are independently integers from 0-10;
  • n is an integer from 1-5.
  • At least one of R 1 and R 2 is alkyl, unsubstituted or substituted with halo-. In another example, both R 1 and R 2 are independently alkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of R 1 and R 2 is methyl. In other embodiments, R 1 and R 2 are methyl.
  • x+y+z is at least 1. In other embodiments of the invention, x+y+z is at least 2. In other embodiments of the invention, x+y+z is 3, 4, 5, 6, 7, 8, 9 or 10.
  • Each occurrence of A, B, C, D or E in a macrocycle or macrocycle precursor of the invention is independently selected.
  • a sequence represented by the formula [A] x when x is 3, encompasses embodiments where the amino acids are not identical, e.g. Gln-Asp-Ala as well as embodiments where the amino acids are identical, e.g. Gln-Gln-Gln. This applies for any value of x, y, or z in the indicated ranges.
  • the peptidomimetic macrocycle of the invention comprises a secondary structure which is a helix and R is —H, allowing intrahelical hydrogen bonding.
  • at least one of A, B, C, D or E is an ⁇ , ⁇ -disubstituted amino acid.
  • 13 is an ⁇ , ⁇ -disubstituted amino acid.
  • at least one of A, B, C, D or E is 2-aminoisobutyric acid.
  • at least one of A, B, C, D or E is
  • the length of the macrocycle-forming linker L as measured from a first C ⁇ to a second C ⁇ is selected to stabilize a desired secondary peptide structure, such as a helix (including a 3 10 helix or ⁇ -helix) formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first C ⁇ to a second C ⁇ .
  • a desired secondary peptide structure such as a helix (including a 3 10 helix or ⁇ -helix) formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first C ⁇ to a second C ⁇ .
  • Peptidomimetic macrocycles of the invention may be prepared by any of a variety of methods known in the art.
  • any of the residues indicated by “X” in Table 1 may be substituted with a residue capable of forming a crosslinker with a second residue in the same molecule or a precursor of such a residue.
  • peptidomimetic macrocycles Various methods to effect formation of peptidomimetic macrocycles are known in the art. For example, the preparation of peptidomimetic macrocycles of Formula I is described in Schafmeister et al., J. Am. Chem. Soc. 122:5891-5892 (2000); Schafffle & Verdine, J. Am. Chem. Soc. 122:5891 (2005); Walensky et al, Science 305:1466-1470 (2004); and U.S. Pat. No. 7,192,713.
  • the ⁇ , ⁇ -disubstituted amino acids and amino acid precursors disclosed in the cited references may be employed in synthesis of the peptidomimetic macrocycle precursor polypeptides.
  • the “S5-olefin amino acid” is (S)- ⁇ -(2′-pentenyl) alanine and the “R8 olefin amino acid” is (R)- ⁇ -(2′-octenyl) alanine.
  • the terminal olefins are reacted with a metathesis catalyst, leading to the formation of the peptidomimetic macrocycle.
  • the peptidomimetic macrocycles of the invention are of Formula IV or IVa. Methods for the preparation of such macrocycles are described, for example, in U.S. Pat. No. 7,202,332.
  • the synthesis of these peptidomimetic macrocycles involves a multi-step process that features the synthesis of a peptidomimetic precursor containing an azide moiety and an alkyne moiety; followed by contacting the peptidomimetic precursor with a macrocyclization reagent to generate a triazole-linked peptidomimetic macrocycle.
  • a multi-step process that features the synthesis of a peptidomimetic precursor containing an azide moiety and an alkyne moiety; followed by contacting the peptidomimetic precursor with a macrocyclization reagent to generate a triazole-linked peptidomimetic macrocycle.
  • Macrocycles or macrocycle precursors are synthesized, for example, by solution phase or solid-phase methods, and can contain both naturally-occurring and non-naturally-occurring amino acids. See, for example, Hunt, “The Non-Protein Amino Acids” in Chemistry and Biochemistry of the Amino Acids , edited by
  • an azide is linked to the ⁇ -carbon of a residue and an alkyne is attached to the ⁇ -carbon of another residue.
  • the azide moieties are azido-analogs of amino acids L-lysine, D-lysine, alpha-methyl-L-lysine, alpha-methyl-D-lysine, L-omithine, D-ornithine, alpha-methyl-L-omithine or alpha-methyl-D-ornithine.
  • the alkyne moiety is L-propargylglycine.
  • the alkyne moiety is an amino acid selected from the group consisting of L-propargylglycine, D-propargylglycine, (S)-2-amino-2-methyl-4-pentynoic acid, (R)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-2-methyl-5-hexynoic acid, (R)-2-amino-2-methyl-5-hexynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, (R)-2-amino-2-methyl-6-heptynoic acid, (S)-2-amino-2-methyl-7-octynoic acid, (R)-2-amino-2-methyl-7-octynoic acid, (S)-2-amino-2-methyl-8-nonynoic acid and (R)-2-amino-2-methyl-8-nonynoic acid.
  • the invention provides a method for synthesizing a peptidomimetic macrocycle, the method comprising the steps of contacting a peptidomimetic precursor of Formula V or Formula VI:
  • R 12 is —H when the macrocyclization reagent is a Cu reagent and R 12 is —H or alkyl when the macrocyclization reagent is a Ru reagent; and further wherein said contacting step results in a covalent linkage being formed between the alkyne and azide moiety in Formula III or Formula IV.
  • R 12 may be methyl when the macrocyclization reagent is a Ru reagent.
  • R 1 and R 2 are alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-.
  • both R 1 and R 2 are independently alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-.
  • At least one of A, B, C, D or E is an ⁇ , ⁇ -disubstituted amino acid.
  • B is an ⁇ , ⁇ -disubstituted amino acid.
  • at least one of A, B, C, D or E is 2-aminoisobutyric acid.
  • R 1 and R 2 are alkyl, unsubstituted or substituted with halo-. In another example, both R 1 and R 2 are independently alkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of R 1 and R 2 is methyl. In other embodiments, R 1 and R 2 are methyl.
  • the macrocyclization reagent may be a Cu reagent or a Ru reagent.
  • the peptidomimetic precursor is purified prior to the contacting step.
  • the peptidomimetic macrocycle is purified after the contacting step.
  • the peptidomimetic macrocycle is refolded after the contacting step.
  • the method may be performed in solution, or, alternatively, the method may be performed on a solid support.
  • Also envisioned herein is performing the method of the invention in the presence of a target macromolecule that binds to the peptidomimetic precursor or peptidomimetic macrocycle under conditions that favor said binding.
  • the method is performed in the presence of a target macromolecule that binds preferentially to the peptidomimetic precursor or peptidomimetic macrocycle under conditions that favor said binding.
  • the method may also be applied to synthesize a library of peptidomimetic macrocycles.
  • the alkyne moiety of the peptidomimetic precursor of Formula V or Formula VI is a sidechain of an amino acid selected from the group consisting of L-propargylglycine, D-propargylglycine, (S)-2-amino-2-methyl-4-pentynoic acid, (R)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-2-methyl-5-hexynoic acid, (R)-2-amino-2-methyl-5-hexynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, (R)-2-amino-2-methyl-6-heptynoic acid, (S)-2-amino-2-methyl-7-octynoic acid, (R)-2-amino-2-methyl-7-octynoic acid, (S)-2-amino-2-methyl-8-nonynoic acid, and (R)-2-amino-2-amino
  • the azide moiety of the peptidomimetic precursor of Formula V or Formula VI is a sidechain of an amino acid selected from the group consisting of ⁇ -azido-L-lysine, ⁇ -azido-D-lysine, ⁇ -azido- ⁇ -methyl-L-lysine, ⁇ -azido- ⁇ -methyl-D-lysine, ⁇ -azido- ⁇ -methyl-L-omithine, and ⁇ -azido- ⁇ -methyl-D-ornithine.
  • x+y+z is 2, and A, B and C are independently natural or non-natural amino acids. In other embodiments, x+y+z is 3 or 6, and A, B and C are independently natural or non-natural amino acids.
  • the contacting step is performed in a solvent selected from the group consisting of protic solvent, aqueous solvent, organic solvent, and mixtures thereof.
  • the solvent may be chosen from the group consisting of H 2 O, THF, THF/H 2 O, tBuOH/H 2 O, DMF, DIPEA, CH 3 CN or CH 2 Cl 2 , ClCH 2 CH 2 Cl or a mixture thereof.
  • the solvent may be a solvent which favors helix formation.
  • peptidomimetic macrocycles of the invention have the structure (SEQ ID NOS 59-62, respectively, in order of appearance):
  • influenza PB1 peptidomimetic macrocycles shown above are also identified as SP-8, SP-16, SP-13 and SP-41, respectively.
  • the peptidomimetic macrocycle of the invention has the structure (SEQ ID NOS 63-65, respectively, in order of appearance):
  • peptidomimetic macrocycles of the invention are made, for example, by chemical synthesis methods, such as described in Fields et al., Chapter 3 in Synthetic Peptides: A User's Guide , ed. Grant, W. H. Freeman & Co., New York, N.Y., 1992, p. 77.
  • peptides are synthesized using the automated Merrifield techniques of solid phase synthesis with the amine protected by either tBoc or Fmoc chemistry using side chain protected amino acids on, for example, an automated peptide synthesizer (e.g., Applied Biosystems (Foster City, Calif.), Model 430A, 431, or 433).
  • One manner of producing the peptidomimetic precursors and peptidomimetic macrocycles described herein uses solid phase peptide synthesis (SPPS).
  • SPPS solid phase peptide synthesis
  • the C-terminal amino acid is attached to a cross-linked polystyrene resin via an acid labile bond with a linker molecule.
  • This resin is insoluble in the solvents used for synthesis, making it relatively simple and fast to wash away excess reagents and by-products.
  • the N-terminus is protected with the Fmoc group, which is stable in acid, but removable by base. Side chain functional groups are protected as necessary with base stable, acid labile groups.
  • peptidomimetic precursors are produced, for example, by conjoining individual synthetic peptides using native chemical ligation. Alternatively, the longer synthetic peptides are biosynthesized by well known recombinant DNA and protein expression techniques. Such techniques are provided in well-known standard manuals with detailed protocols.
  • To construct a gene encoding a peptidomimetic precursor of this invention the amino acid sequence is reverse translated to obtain a nucleic acid sequence encoding the amino acid sequence, preferably with codons that are optimum for the organism in which the gene is to be expressed.
  • a synthetic gene is made, typically by synthesizing oligonucleotides which encode the peptide and any regulatory elements, if necessary.
  • the synthetic gene is inserted in a suitable cloning vector and transfected into a host cell. The peptide is then expressed under suitable conditions appropriate for the selected expression system and host.
  • the peptide is purified and characterized by standard methods.
  • the peptidomimetic precursors are made, for example, in a high-throughput, combinatorial fashion using, for example, a high-throughput polychannel combinatorial synthesizer (e.g., Thuramed TETRAS multichannel peptide synthesizer from CreoSalus, Louisville, Ky. or Model Apex 396 multichannel peptide synthesizer from AAPPTEC, Inc., Louisville, Ky.).
  • a high-throughput polychannel combinatorial synthesizer e.g., Thuramed TETRAS multichannel peptide synthesizer from CreoSalus, Louisville, Ky. or Model Apex 396 multichannel peptide synthesizer from AAPPTEC, Inc., Louisville, Ky.
  • each R 1 , R 2 , R 7 and R 8 is —H; each L 1 is —(CH 2 ) 4 —; and each L 2 is —(CH 2 )—.
  • R 1 , R 2 , R 7 , R 8 , L 1 and L 2 can be independently selected from the various structures disclosed herein.
  • Synthetic Scheme 1 describes the preparation of several compounds of the invention.
  • Ni(II) complexes of Schiff bases derived from the chiral auxiliary (S)-2-[N—(N′-benzylpropyl)amino]benzophenone (BPB) and amino acids such as glycine or alanine are prepared as described in Belokon et al. (1998), Tetrahedron Asymm. 9:4249-4252.
  • the resulting complexes are subsequently reacted with alkylating reagents comprising an azido or alkynyl moiety to yield enantiomerically enriched compounds of the invention. If desired, the resulting compounds can be protected for use in peptide synthesis.
  • the peptidomimetic precursor contains an azide moiety and an alkyne moiety and is synthesized by solution-phase or solid-phase peptide synthesis (SPPS) using the commercially available amino acid N- ⁇ -Fmoc-L-propargylglycine and the N- ⁇ -Fmoc-protected forms of the amino acids (S)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-6-heptynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, N-methyl- ⁇ -azido-L-lysine, and N-methyl- ⁇ -azido-D-lysine.
  • SPPS solution-phase or solid-phase peptide synthesis
  • the peptidomimetic precursor is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA).
  • the peptidomimetic precursor is reacted as a crude mixture or is purified prior to reaction with a macrocyclization reagent such as a Cu(I) in organic or aqueous solutions (Rostovtsev et al. (2002), Angew. Chem. Int. Ed. 41:2596-2599; Tornoe et al. (2002), J. Org. Chem. 67:3057-3064; Deiters et al. (2003), J. Am. Chem. Soc.
  • the triazole forming reaction is performed under conditions that favor helix formation.
  • the macrocyclization step is performed in a solvent chosen from the group consisting of H 2 O, THF, CH 3 CN, DMF, DIPEA, tBuOH or a mixture thereof.
  • the macrocyclization step is performed in DMF.
  • the macrocyclization step is performed in a buffered aqueous or partially aqueous solvent.
  • the peptidomimetic precursor contains an azide moiety and an alkyne moiety and is synthesized by solid-phase peptide synthesis (SPPS) using the commercially available amino acid N- ⁇ -Fmoc-L-propargylglycine and the N- ⁇ -Fmoc-protected forms of the amino acids (S)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-6-heptynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, N-methyl- ⁇ -azido-L-lysine, and N-methyl- ⁇ -azido-D-lysine.
  • SPPS solid-phase peptide synthesis
  • the peptidomimetic precursor is reacted with a macrocyclization reagent such as a Cu(I) reagent on the resin as a crude mixture
  • a macrocyclization reagent such as a Cu(I) reagent
  • the resultant triazole-containing peptidomimetic macrocycle is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA).
  • the macrocyclization step is performed in a solvent chosen from the group consisting of CH 2 Cl 2 , ClCH 2 CH 2 Cl, DMF, THF, NMP, DIPEA, 2,6-lutidine, pyridine, DMSO, H 2 O or a mixture thereof.
  • the macrocyclization step is performed in a buffered aqueous or partially aqueous solvent.
  • the peptidomimetic precursor contains an azide moiety and an alkyne moiety and is synthesized by solution-phase or solid-phase peptide synthesis (SPPS) using the commercially available amino acid N- ⁇ -Fmoc-L-propargylglycine and the N- ⁇ -Fmoc-protected forms of the amino acids (S)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-6-heptynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, N-methyl- ⁇ -azido-L-lysine, and N-methyl- ⁇ -azido-D-lysine.
  • SPPS solution-phase or solid-phase peptide synthesis
  • the peptidomimetic precursor is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA).
  • the peptidomimetic precursor is reacted as a crude mixture or is purified prior to reaction with a macrocyclization reagent such as a Ru(II) reagents, for example Cp*RuCl(PPh 3 ) 2 or [Cp*RuCl] 4 (Rasmussen et al. (2007), Org. Lett. 9:5337-5339; Zhang et al. (2005), J. Am. Chem. Soc. 127:15998-15999).
  • the macrocyclization step is performed in a solvent chosen from the group consisting of DMF, CH 3 CN and THF.
  • the peptidomimetic precursor contains an azide moiety and an alkyne moiety and is synthesized by solid-phase peptide synthesis (SPPS) using the commercially available amino acid N- ⁇ -Fmoc-L-propargylglycine and the N- ⁇ -Fmoc-protected forms of the amino acids (S)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-6-heptynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, N-methyl- ⁇ -azido-L-lysine, and N-methyl- ⁇ -azido-D-lysine.
  • SPPS solid-phase peptide synthesis
  • the peptidomimetic precursor is reacted with a macrocyclization reagent such as a Ru(H) reagent on the resin as a crude mixture.
  • a macrocyclization reagent such as a Ru(H) reagent on the resin as a crude mixture.
  • the reagent can be Cp*RuCl(PPh 3 ) 2 or [Cp*RuCl] 4 (Rasmussen et al. (2007), Org. Lett. 9:5337-5339; Zhang et al. (2005), J. Am. Chem. Soc. 127:15998-15999).
  • the macrocyclization step is performed in a solvent chosen from the group consisting of CH 2 Cl 2 , ClCH 2 CH 2 Cl, CH 3 CN, DMF, and THF.
  • the present invention contemplates the use of non-naturally-occurring amino acids and amino acid analogs in the synthesis of the peptidomimetic macrocycles described herein.
  • Any amino acid or amino acid analog amenable to the synthetic methods employed for the synthesis of stable triazole containing peptidomimetic macrocycles can be used in the present invention.
  • L-propargylglycine is contemplated as a useful amino acid in the present invention.
  • other alkyne-containing amino acids that contain a different amino acid side chain are also useful in the invention.
  • L-propargylglycine contains one methylene unit between the ⁇ -carbon of the amino acid and the alkyne of the amino acid side chain.
  • the invention also contemplates the use of amino acids with multiple methylene units between the ⁇ -carbon and the alkyne.
  • the azido-analogs of amino acids L-lysine, D-lysine, alpha-methyl-L-lysine, and alpha-methyl-D-lysine are contemplated as useful amino acids in the present invention.
  • other terminal azide amino acids that contain a different amino acid side chain are also useful in the invention.
  • the azido-analog of L-lysine contains four methylene units between the ⁇ -carbon of the amino acid and the terminal azide of the amino acid side chain.
  • the invention also contemplates the use of amino acids with fewer than or greater than four methylene units between the ⁇ -carbon and the terminal azide. Table 2 shows some amino acids useful in the preparation of peptidomimetic macrocycles of the invention.
  • Table 2 shows exemplary amino acids useful in the preparation of peptidomimetic macrocycles of the invention.
  • the amino acids and amino acid analogs are of the D-configuration. In other embodiments they are of the L-configuration. In some embodiments, some of the amino acids and amino acid analogs contained in the peptidomimetic are of the D-configuration while some of the amino acids and amino acid analogs are of the L-configuration. In some embodiments the amino acid analogs are ⁇ , ⁇ -disubstituted, such as ⁇ -methyl-L-propargylglycine, ⁇ -methyl-D-propargylglycine, ⁇ -azido-alpha-methyl-L-lysine, and ⁇ -azido-alpha-methyl-D-lysine.
  • amino acid analogs are N-alkylated, e.g., N-methyl-L-propargylglycine, N-methyl-D-propargylglycine, N-methyl- ⁇ -azido-L-lysine, and N-methyl- ⁇ -azido-D-lysine.
  • the —NH moiety of the amino acid is protected using a protecting group, including without limitation -Fmoc and -Boc. In other embodiments, the amino acid is not protected prior to synthesis of the peptidomimetic macrocycle.
  • peptidomimetic macrocycles of Formula III are synthesized.
  • the preparation of such macrocycles is described, for example, in U.S. application Ser. No. 11/957,325, filed on Dec. 17, 2007.
  • the following synthetic schemes describe the preparation of such compounds.
  • the illustrative schemes depict amino acid analogs derived from L- or D-cysteine, in which L 1 and L 3 are both —(CH 2 )—.
  • L 1 and L 3 can be independently selected from the various structures disclosed herein.
  • the peptidomimetic precursor contains two —SH moieties and is synthesized by solid-phase peptide synthesis (SPPS) using commercially available N- ⁇ -Fmoc amino acids such as N- ⁇ -Fmoc-S-trityl-L-cysteine or N- ⁇ -Fmoc-S-trityl-D-cysteine.
  • SPPS solid-phase peptide synthesis
  • Alpha-methylated versions of D-cysteine or L-cysteine are generated by known methods (Seebach et al. (1996), Angew. Chem. Int. Ed. Engl.
  • N- ⁇ -Fmoc-S-trityl monomers by known methods (“ Bioorganic Chemistry: Peptides and Proteins ”, Oxford University Press, New York: 1998, the entire contents of which are incorporated herein by reference).
  • the precursor peptidomimetic is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA).
  • the precursor peptidomimetic is reacted as a crude mixture or is purified prior to reaction with X-L 2 -Y in organic or aqueous solutions.
  • the alkylation reaction is performed under dilute conditions (i.e.
  • the alkylation reaction is performed in organic solutions such as liquid NH 3 (Mosberg et al. (1985), J. Am. Chem. Soc. 107:2986-2987; Szewczuk et al. (1992), Int. J. Peptide Protein Res. 40: 233-242), NH 3 /MeOH, or NH 3 /DMF (Or et al. (1991), J. Org. Chem. 56:3146-3149).
  • the alkylation is performed in an aqueous solution such as 6M guanidinium HCL, pH 8 (Brunel et al. (2005), Chem. Commun. (20):2552-2554).
  • the solvent used for the alkylation reaction is DMF or
  • the precursor peptidomimetic contains two or more —SH moieties, of which two are specially protected to allow their selective deprotection and subsequent alkylation for macrocycle formation.
  • the precursor peptidomimetic is synthesized by solid-phase peptide synthesis (SPPS) using commercially available N- ⁇ -Fmoc amino acids such as N- ⁇ -Fmoc-S-p-methoxytrityl-L-cysteine or N- ⁇ -Fmoc-S-p-methoxytrityl-D-cysteine.
  • SPPS solid-phase peptide synthesis
  • Alpha-methylated versions of D-cysteine or L-cysteine are generated by known methods (Seebach et al. (1996), Angew. Chem. Int. Ed.
  • the alkylation reaction is performed in organic solutions such as liquid NH 3 (Mosberg et al. (1985), J. Am. Chem. Soc. 107:2986-2987; Szewczuk et al. (1992), Int. J. Peptide Protein Res. 40:233-242), NH 3 /MeOH or NH 3 /DMF (Or et al. (1991), J. Org. Chem. 56:3146-3149).
  • the alkylation reaction is performed in DMF or dichloroethane.
  • the peptidomimetic macrocycle is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA).
  • the peptidomimetic precursor contains two or more —SH moieties, of which two are specially protected to allow their selective deprotection and subsequent alkylation for macrocycle formation.
  • the peptidomimetic precursor is synthesized by solid-phase peptide synthesis (SPPS) using commercially available N- ⁇ -Fmoc amino acids such as N- ⁇ -Fmoc-S-p-methoxytrityl-L-cysteine, N- ⁇ -Fmoc-S-p-methoxytrityl-D-cysteine, N- ⁇ -Fmoc-S—S-t-butyl-L-cysteine, and N- ⁇ -Fmoc-S—S-t-butyl-D-cysteine.
  • SPPS solid-phase peptide synthesis
  • Alpha-methylated versions of D-cysteine or L-cysteine are generated by known methods (Secbach et al. (1996), Angew. Chem. Int. Ed. Engl. 35:2708-2748, and references therein) and then converted to the appropriately protected N- ⁇ -Fmoc-S-p-methoxytrityl or N- ⁇ -Fmoc-S—S-t-butyl monomers by known methods ( Bioorganic Chemistry: Peptides and Proteins , Oxford University Press, New York: 1998, the entire contents of which are incorporated herein by reference).
  • the S—S-tButyl protecting group of the peptidomimetic precursor is selectively cleaved by known conditions (e.g., 20% 2-mercaptoethanol in DMF, reference: Gauß et al. (2005), J. Comb. Chem. 7:174-177).
  • the precursor peptidomimetic is then reacted on the resin with a molar excess of X-L 2 -Y in an organic solution.
  • the reaction takes place in the presence of a hindered base such as diisopropylethylamine.
  • the Mmt protecting group of the peptidomimetic precursor is then selectively cleaved by standard conditions (e.g., mild acid such as 1% TFA in DCM).
  • the peptidomimetic precursor is then cyclized on the resin by treatment with a hindered base in organic solutions.
  • the alkylation reaction is performed in organic solutions such as NH 3 /MeOH or NH 3 /DMF (Or et al. (1991), J. Org. Chem. 56:3146-3149).
  • the peptidomimetic macrocycle is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA).
  • the peptidomimetic precursor contains two L-cysteine moieties.
  • the peptidomimetic precursor is synthesized by known biological expression systems in living cells or by known in vitro, cell-free, expression methods.
  • the precursor peptidomimetic is reacted as a crude mixture or is purified prior to reaction with X-L2-Y in organic or aqueous solutions.
  • the alkylation reaction is performed under dilute conditions (i.e. 0.15 mmol/L) to favor macrocyclization and to avoid polymerization.
  • the alkylation reaction is performed in organic solutions such as liquid NH 3 (Mosberg et al. (1985), J. Am. Chem. Soc.
  • the alkylation is performed in an aqueous solution such as 6M guanidinium HCL, pH 8 (Brunel et al. (2005), Chem. Commun. (20):2552-2554). In other embodiments, the alkylation is performed in DMF or dichloroethane.
  • the alkylation is performed in non-denaturing aqueous solutions, and in yet another embodiment the alkylation is performed under conditions that favor helical structure formation. In yet another embodiment, the alkylation is performed under conditions that favor the binding of the precursor peptidomimetic to another protein, so as to induce the formation of the bound helical conformation during the alkylation.
  • X and Y are envisioned which are suitable for reacting with thiol groups.
  • each X or Y is independently be selected from the general category shown in Table 5.
  • X and Y are halides such as —Cl, —Br or —I.
  • Any of the macrocycle-forming linkers described herein may be used in any combination with any of the sequences shown in Tables 1-4 and also with any of the R-substituents indicated herein.
  • the present invention contemplates the use of both naturally-occurring and non-naturally-occurring amino acids and amino acid analogs in the synthesis of the peptidomimetic macrocycles of Formula (III).
  • Any amino acid or amino acid analog amenable to the synthetic methods employed for the synthesis of stable bis-sulfhydryl containing peptidomimetic macrocycles can be used in the present invention.
  • cysteine is contemplated as a useful amino acid in the present invention.
  • sulfur containing amino acids other than cysteine that contain a different amino acid side chain are also useful.
  • cysteine contains one methylene unit between the ⁇ -carbon of the amino acid and the terminal —SH of the amino acid side chain.
  • the invention also contemplates the use of amino acids with multiple methylene units between the ⁇ -carbon and the terminal —SH.
  • Non-limiting examples include ⁇ -methyl-L-homocysteine and ⁇ -methyl-D-homocysteine.
  • the amino acids and amino acid analogs are of the D-configuration. In other embodiments they are of the L-configuration.
  • some of the amino acids and amino acid analogs contained in the peptidomimetic are of the D-configuration while some of the amino acids and amino acid analogs are of the L-configuration.
  • the amino acid analogs are ⁇ , ⁇ -disubstituted, such as ⁇ -methyl-L-cysteine and ⁇ -methyl-D-cysteine.
  • the invention includes macrocycles in which macrocycle-forming linkers are used to link two or more —SH moieties in the peptidomimetic precursors to form the peptidomimetic macrocycles of the invention.
  • the macrocycle-forming linkers impart conformational rigidity, increased metabolic stability and/or increased cell penetrability.
  • the macrocycle-forming linkages stabilize a helical secondary structure of the peptidomimetic macrocyles.
  • the macrocycle-forming linkers are of the formula X-L 2 -Y, wherein both X and Y are the same or different moieties, as defined above.
  • Both X and Y have the chemical characteristics that allow one macrocycle-forming linker -L 2 - to bis alkylate the bis-sulfhydryl containing peptidomimetic precursor.
  • the linker -L 2 - includes alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, or heterocycloarylene, or —R 4 —K—R 4 —, all of which can be optionally substituted with an R 5 group, as defined above.
  • one to three carbon atoms within the macrocycle-forming linkers other than the carbons attached to the —SH of the sulfhydryl containing amino acid are optionally substituted with a heteroatom such as N, S or O.
  • the L 2 component of the macrocycle-forming linker X-L 2 -Y may be varied in length depending on, among other things, the distance between the positions of the two amino acid analogs used to form the peptidomimetic macrocycle. Furthermore, as the lengths of L 1 and/or L 3 components of the macrocycle-forming linker are varied, the length of L 2 can also be varied in order to create a linker of appropriate overall length for forming a stable peptidomimetic macrocycle. For example, if the amino acid analogs used are varied by adding an additional methylene unit to each of L 1 and L 3 , the length of L 2 are decreased in length by the equivalent of approximately two methylene units to compensate for the increased lengths of L 1 and L 3 .
  • L 2 is an alkylene group of the formula —(CH 2 ) n —, where n is an integer between about 1 and about 15. For example, n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. In other embodiments, L 2 is an alkenylene group. In still other embodiments, L 2 is an aryl group.
  • Table 4 shows additional embodiments of X-L 2 -Y groups.
  • Additional methods of forming peptidomimetic macrocycles which are envisioned as suitable to perform the present invention include those disclosed by Mustapa, M. Firouz Mohd et al., J. Org. Chem (2003), 68, pp. 8193-8198; Yang, Bin et al. Bioorg Med. Chem. Left. (2004), 14, pp. 1403-1406; U.S. Pat. No. 5,364,851; U.S. Pat. No. 5,446,128; U.S. Pat. No. 5,824,483; U.S. Pat. No. 6,713,280; and U.S. Pat. No. 7,202,332.
  • aminoacid precursors are used containing an additional substituent R— at the alpha position.
  • Such aminoacids are incorporated into the macrocycle precursor at the desired positions, which may be at the positions where the crosslinker is substituted or, alternatively, elsewhere in the sequence of the macrocycle precursor. Cyclization of the precursor is then effected according to the indicated method.
  • the helical macrocycle of the invention comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more Aib substitutions.
  • peptidomimetic macrocycles of the invention are assayed, for example, by using the methods described below.
  • a peptidomimetic macrocycle of the invention has improved biological properties relative to a corresponding polypeptide lacking the substituents described herein.
  • the secondary structure of polypeptides with helical domains will reach a dynamic equilibrium between random coil structures and helical structures, often expressed as a “percent helicity”.
  • unmodified helical domains may be predominantly random coils in solution, with helical content usually under 25%.
  • Peptidomimetic macrocycles with optimized linkers possess, for example, a helicity that is at least two-fold greater than that of a corresponding uncrosslinked polypeptide.
  • macrocycles of the invention will possess a helicity of greater than 50%.
  • an aqueous solution e.g.
  • Circular dichroism (CD) spectra are obtained on a spectropolarimeter (e.g., Jasco J-710) using standard measurement parameters (e.g. temperature, 20° C.; wavelength, 190-260 nm; step resolution, 0.5 nm; speed, 20 nm/sec; accumulations, 10; response, 1 sec; bandwidth, 1 nm; path length, 0.1 cm).
  • the helical content of each peptide is calculated by dividing the mean residue ellipticity (e.g. [ ⁇ ]222obs) by the reported value for a model helical decapeptide (Yang et al. (1986), Methods Enzymol. 130:208)).
  • a peptidomimetic macrocycle of the invention comprising a secondary structure such as a helix exhibits, for example, a higher melting temperature than a corresponding uncrosslinked polypeptide.
  • peptidomimetic macrocycles of the invention exhibit Tm of >60° C. representing a highly stable structure in aqueous solutions.
  • Tm is determined by measuring the change in ellipticity over a temperature range (e.g.
  • spectropolarimeter e.g., Jasco J-710
  • standard parameters e.g. wavelength 222 nm; step resolution, 0.5 nm; speed, 20 nm/sec; accumulations, 10; response, 1 sec; bandwidth, 1 nm; temperature increase rate: 1° C./min; path length, 0.1 cm.
  • the amide bond of the peptide backbone is susceptible to hydrolysis by proteases, thereby rendering peptidic compounds vulnerable to rapid degradation in vivo. Peptide helix formation, however, typically buries the amide backbone and therefore may shield it from proteolytic cleavage.
  • the peptidomimetic macrocycles of the present invention may be subjected to in vitro trypsin proteolysis to assess for any change in degradation rate compared to a corresponding uncrosslinked polypeptide.
  • the peptidomimetic macrocycle and a corresponding uncrosslinked polypeptide are incubated with trypsin agarose and the reactions quenched at various time points by centrifugation and subsequent HPLC injection to quantitate the residual substrate by ultraviolet absorption at 280 nm.
  • the peptidomimetic macrocycle and peptidomimetic precursor (5 mcg) are incubated with trypsin agarosc (Pierce) (S/E ⁇ 125) for 0, 10, 20, 90, and 180 minutes. Reactions are quenched by tabletop centrifugation at high speed; remaining substrate in the isolated supernatant is quantified by HPLC-based peak detection at 280 nm.
  • Peptidomimetic macrocycles with optimized linkers possess, for example, an ex vivo half-life that is at least two-fold greater than that of a corresponding uncrosslinked polypeptide, and possess an ex vivo half-life of 12 hours or more.
  • assays may be used. For example, a peptidomimetic macrocycle and a corresponding uncrosslinked polypeptide (2 meg) are incubated with fresh mouse, rat and/or human serum (2 mL) at 37° C. for 0, 1, 2, 4, 8, and 24 hours.
  • the samples are extracted by transferring 100 ⁇ l of sera to 2 ml centrifuge tubes followed by the addition of 10 ⁇ L of 50% formic acid and 500 ⁇ L acetonitrile and centrifugation at 14,000 RPM for 10 min at 4 ⁇ 2° C. The supernatants are then transferred to fresh 2 ml tubes and evaporated on Turbovap under N 2 ⁇ 10 psi, 37° C. The samples are reconstituted in 100 ⁇ L, of 50:50 acetonitrile:water and submitted to LC-MS/MS analysis.
  • FPA fluorescence polarization assay
  • fluoresceinated peptidomimetic macrocycles (25 nM) are incubated with the acceptor protein (25-1000 nM) in binding buffer (140 mM NaCl, 50 mM Tris-HCL, pH 7.4) for 30 minutes at room temperature. Binding activity is measured, for example, by fluorescence polarization on a luminescence spectrophotometer (e.g. Perkin-Elmer LS50B). Kd values may be determined by nonlinear regression analysis using, for example, Graphpad Prism software (GraphPad Software, Inc., San Diego, Calif.).
  • a peptidomimetic macrocycle of the invention shows, in some instances, similar or lower Kd than a corresponding uncrosslinked polypeptide.
  • a fluorescence polarization assay utilizing a fluoresceinated peptidomimetic macrocycle derived from a peptidomimetic precursor sequence is used, for example.
  • the FPA technique measures the molecular orientation and mobility using polarized light and fluorescent tracer.
  • fluorescent tracers e.g., FITC
  • FITC-labeled peptides bound to a large protein When excited with polarized light, fluorescent tracers (e.g., FITC) attached to molecules with high apparent molecular weights (e.g. FITC-labeled peptides bound to a large protein) emit higher levels of polarized fluorescence due to their slower rates of rotation as compared to fluorescent tracers attached to smaller molecules (e.g. FITC-labeled peptides that are free in solution).
  • a compound that antagonizes the interaction between the fluoresceinated peptidomimetic macrocycle and an acceptor protein will be detected in a competitive binding FPA experiment
  • putative antagonist compounds (1 nM to 1 mM) and a fluoresceinated peptidomimetic macrocycle (25 nM) are incubated with the acceptor protein (50 nM) in binding buffer (140 mM NaCl, 50 mM Tris-HCL, pH 7.4) for 30 minutes at room temperature.
  • Antagonist binding activity is measured, for example, by fluorescence polarization on a luminescence spectrophotometer (e.g. Perkin-Elmer LS50B).
  • Kd values may be determined by nonlinear regression analysis using, for example, Graphpad Prism software (GraphPad Software, Inc., San Diego, Calif.).
  • Any class of molecule such as small organic molecules, peptides, oligonucleotides or proteins can be examined as putative antagonists in this assay.
  • FITC-labeled fluoresceinated compounds
  • lysis buffer 50 mM Tris [pH 7.6], 150 mM NaCl, 1% CHAPS and protease inhibitor cocktail
  • Extracts are centrifuged at 14,000 rpm for 15 minutes and supernatants collected and incubated with 10 ⁇ l goat anti-FITC antibody for 2 hrs, rotating at 4° C. followed by further 2 hrs incubation at 4° C. with protein A/G Sepharose (50 ⁇ l of 50% bead slurry). After quick centrifugation, the pellets are washed in lysis buffer containing increasing salt concentration (e.g., 150, 300, 500 mM). The beads are then re-equilibrated at 150 mM NaCl before addition of SDS-containing sample buffer and boiling.
  • increasing salt concentration e.g. 150, 300, 500 mM
  • the supernatants are optionally electrophoresed using 4%-12% gradient Bis-Tris gels followed by transfer into Immobilon-P membranes. After blocking, blots are optionally incubated with an antibody that detects FITC and also with one or more antibodies that detect proteins that bind to the peptidomimetic macrocycle.
  • a peptidomimetic macrocycle is, for example, more cell penetrable compared to a corresponding uncrosslinked macrocycle.
  • Peptidomimetic macrocycles with optimized linkers possess, for example, cell penetrability that is at least two-fold greater than a corresponding uncrosslinked macrocycle, and often 20% or more of the applied peptidomimetic macrocycle will be observed to have penetrated the cell after 4 hours.
  • peptidomimetic macrocycles and corresponding uncrosslinked macrocycle To measure the cell penetrability of peptidomimetic macrocycles and corresponding uncrosslinked macrocycle, intact cells are incubated with fluoresceinated peptidomimetic macrocycles or corresponding uncrosslinked macrocycle (10 ⁇ M) for 4 hrs in serum free media at 37° C., washed twice with media and incubated with trypsin (0.25%) for 10 min at 37° C. The cells are washed again and resuspended in PBS. Cellular fluorescence is analyzed, for example, by using either a FACS Calibur flow cytometer or Cellomics' KineticScan® HCS Reader.
  • the compounds are, for example, administered to mice and/or rats by IV, IP, PO or inhalation routes at concentrations ranging from 0.1 to 50 mg/kg and blood specimens withdrawn at 0′, 5′, 15′, 30′, 1 hr, 4 hrs, 8 hrs and 24 hours post-injection. Levels of intact compound in 25 ⁇ L of fresh scrum are then measured by LC-MS/MS as above.
  • This influenza antiviral evaluation assay examines the effects of compounds at designated dose-response concentrations. See also Arthur, J. W., W. Severson, D. L. Arthur, L. Rasmussen, E. L. White, and C. B. Jonsson, Antiviral Res, 2007. 73(1): p. 50-9. Malin Darby canine kidney (MDCK) cells are used in the assay to test the efficacy of the compounds in preventing the cytopathic effect (CPE) induced by influenza infection. Either Ribavirin or Tamiflu is included in each run as a positive control compound.
  • MDCK Malin Darby canine kidney
  • Subconfluent cultures of MDCK cells are plated into 96-well plates for the analysis of cell viability (cytotoxicity) and antiviral activity (CPE). Drugs are added to the cells 24 hours later. At a designated time, the CPE wells also receive 100 tissue culture infectious doses (100 TCID 50 s) of titered influenza virus. 72 hours later the cell viability is determined.
  • ferrets Mustela putorius furo
  • ferrets Mustela putorius furo
  • these animals have been widely used as a model for influenza virus pathogenesis and immunity studies.
  • Sec Sidwell, R. W. and D. F. Smec Antiviral Res, 2000. 48(1): p. 1-16; and Colacino, J. M., D. C. DeLong, J. R. Nelson, W. A. Spitzer, J. Tang, F. Victor, and C. Y. Wu, Antimicrob Agents Chemother, 1990. 34(11): p. 2156-63.
  • Ferrets are also the model of choice for the study of avian influenza virus H5N1 pathogenesis in mammals. See also Zitzow, L. A., T. Rowe, T. Morken, W.-J. Shieh, S. Zaki, and J. M. Katz, Pathogenesis of Avian Influenza A (H5N1) Viruses in Ferrets. 2002. p. 4420-4429.
  • the activities of the PB1 Stapled Peptides can be compared to Ribavirin or Oseltamivir as a positive control.
  • young adult male or female ferrets (five ferrets for each treatment group) that are serologically negative by hemagglutination inhibition assay for currently circulating human influenza A or B viruses are quarantined at least 4 days prior to infection in a BSL-3+ animal holding area, where they are housed in cages contained in bioclean portable laminar flow clean room enclosures (Lab Products, Seaford, Del.). Prior to infection, baseline temperatures are measured twice daily for at least 3 days.
  • Ferrets are anesthetized with ketamine (25 mg/kg), xylazine (2 mg/kg), and atropine (0.05 mg/kg) by the intramuscular route and infected intranasally (i.n.) with virus/ml in phosphate-buffered saline (PBS) delivered to the nostrils.
  • PBS phosphate-buffered saline
  • Control animals are mock-infected with an equivalent dilution (1:30) of noninfectious allantoic fluid.
  • Stapled Peptides are administered i.v. or i.p. one hour after virus infection.
  • Temperatures are measured twice daily using either a rectal thermometer or a subcutaneous implantable temperature transponder (BioMedic Data Systems, Inc., Seaford, Del.) with pre-infection values averaged to obtain a baseline temperature for each ferret. The change in temperature (in degrees Celsius) is calculated at each time point for each animal. Clinical signs of sneezing (before anesthesia), inappetence, dyspnea, and level of activity are assessed. A scoring system is also used to assess the activity level, and based on the daily scores for each animal in a group a relative inactivity index will be calculated. Rectal temperature and activity scores are used to assess the severity of influenza infection and the ability of Stapled Peptides to prevent flu symptoms.
  • Bimolecular Fluorescence Complementation may be used to assay the compounds of the invention.
  • N- and C-terminal fragments of fluorescent proteins e.g. GFP or its derivatives
  • the two non-functional halves of the fluorophore following the expression in cells, are brought into close proximity as a result of the specific protein interactions, which initiates folding of the fragments into an active protein and results in a detectable fluorescent signal at the site of the protein-protein complex.
  • the specific interaction between PB1 and PA subunits can be visualized, quantified and localized within live cells.
  • the BiFC signal will be reduced, indicative of the presence of potential inhibitors targeting the assembly of PB1-PA complex. See Hemerka et. al., J. Virol. 2009, 3944-3955.
  • the peptidomimetic macrocycles of the invention also include pharmaceutically acceptable derivatives or prodrugs thereof.
  • a “pharmaceutically acceptable derivative” means any pharmaceutically acceptable salt, ester, salt of an ester, pro-drug or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention.
  • Particularly favored pharmaceutically acceptable derivatives are those that increase the bioavailability of the compounds of the invention when administered to a mammal (e.g., by increasing absorption into the blood of an orally administered compound) or which increases delivery of the active compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • Some pharmaceutically acceptable derivatives include a chemical group which increases aqueous solubility or active transport across the gastrointestinal mucosa.
  • the peptidomimetic macrocycles of the invention are modified by covalently or non-covalently joining appropriate functional groups to enhance selective biological properties.
  • modifications include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism, and alter rate of excretion.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acid salts include acetate, adipate, benzoate, benzenesulfonate, butyrate, citrate, digluconate, dodecylsulfate, formate, fumarate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, palmoate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, tosylate and undecanoate.
  • Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N
  • pharmaceutically acceptable carriers include either solid or liquid carriers.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules.
  • a solid carrier can be one or more substances, which also acts as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. Details on techniques for formulation and administration are well described in the scientific and patent literature, see, e.g., the latest edition of Remington's Pharmaceutical Sciences, Maack Publishing Co, Easton Pa.
  • the carrier is a finely divided solid, which is in a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
  • Suitable solid excipients are carbohydrate or protein fillers include, but are not limited to sugars, including lactose, sucrose, mannitol, or sorbitol; starch from corn, wheat, rice, potato, or other plants; cellulose such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; and gums including arabic and tragacanth; as well as proteins such as gelatin and collagen.
  • disintegrating or solubilizing agents are added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate.
  • Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water/propylene glycol solutions.
  • liquid preparations can be formulated in solution in aqueous polyethylene glycol solution.
  • the pharmaceutical preparation is preferably in unit dosage form.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, cachet, or lozenge itself; or it can be the appropriate number of any of these in packaged form.
  • compositions of this invention comprise a combination of a peptidomimetic macrocycle and one or more additional therapeutic or prophylactic agents
  • both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen.
  • the additional agents are administered separately, as part of a multiple dose regimen, from the compounds of this invention.
  • those agents are part of a single dosage form, mixed together with the compounds of this invention in a single composition.
  • the invention discloses peptidomimetic macrocycles useful in the treatment of viral disorders.
  • peptidomimetic macrocycles derived from the PB1 helix sequence, or peptidomimetic macrocycles that bind selectively to the PB1 peptide binding site of the PA protein may selectively inhibit influenza RNA-dependent RNA polymerases.
  • Peptidomimetic macrocycles derived from the PB2 helix sequence, or peptidomimetic macrocycles that bind selectively to the PB2 peptide binding site of the PB1 protein may selectively inhibit influenza RNA-dependent RNA polymerases.
  • such peptidomimetic macrocycles When administered within a therapeutic window after infection, such peptidomimetic macrocycles may reduce the severity or duration of an influenza infection. When administered prophylactically, such peptidomimetic macrocycles may prevent infection by influenza viruses and thereby decrease the spread of influenza and reduce large-scale epidemics.
  • the present invention provides novel peptidomimetic macrocycles that are useful in competitive binding assays to identify agents which bind to the natural ligand(s) of the proteins or peptides upon which the peptidomimetic macrocycles am modeled.
  • labeled peptidomimetic macrocycles based on PB1 can be used in a PA binding assay along with small molecules that competitively bind to PA.
  • Competitive binding studies allow for rapid in vitro evaluation and determination of drug candidates specific for the PB1/PA system. Such binding studies may be performed with any of the peptidomimetic macrocycles disclosed herein and their binding partners.
  • the present invention provides for both prophylactic and therapeutic methods of treating a subject infected with, at risk of, or susceptible to an influenza virus. These methods comprise administering an effective amount of a compound of the invention to a warm blooded animal, including a human. Tn some embodiments, the administration of the compounds of the present invention prevents the proliferation or transmission of an influenza virus.
  • treatment is defined as the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease or the predisposition toward disease.
  • peptidomimetic macrocycles of the invention are used to treat diseases induced by influenza viruses.
  • influenza virus Like other viruses, the replication of influenza virus involves six phases; transmission, entry, replication, biosynthesis, assembly, and exit. Entry occurs by endocytosis, replication and vRNP assembly takes place in the nucleus, and the virus buds from the plasma membrane. In the infected patient, the virus targets airway epithelial cells.
  • the methods described herein are also useful for development and/or identification of agents for the treatment of infections caused by viruses such as Abelson leukemia virus, Abelson murine leukemia virus, Abelson's virus, Acute laryngotracheobronchitis virus, Sydney River virus, Adeno associated virus group, Adenovirus, African horse sickness virus, African swine fever virus, AIDS virus, Aleutian mink disease parvovirus, Alpharetrovirus, Alphavirus, ALV related virus, Amapari virus, Aphthovirus, Aquareovirus, Arbovirus, Arbovirus C, arbovirus group A, arbovirus group B, Arenavirus group, Argentine hemorrhagic fever virus, Argentine hemorrhagic fever virus, Arterivirus, Astrovirus, Ateline herpesvirus group, Aujezky's disease virus, Aura virus, Ausduk disease virus, Australian bat lyssavirus, Aviadenovirus, avian erythroblastosis virus,
  • influenza virus follows a stepwise, endocytic entry program with elements shared with other viruses such as alpha- and rhabdoviruses (Marsh and Helenius 1989; Whittaker 2006).
  • the steps include: 1) Initial attachment to sialic acid containing glycoconjugates receptors on the cell surface; 2) signaling induced by the virus particle; 3) endocytosis by clathrin-dependent and clathrin-independent cellular mechanism; 4) acid-induced, hemaglutinin (HA)-mediated penetration from late endosomes; 5) acid-activated, M2 and matrix protein (M1) dependent uncoating of the capsid; and, 6) intra-cytosolic transport and nuclear import of vRNPs.
  • These steps depend on assistance from the host cell in the form of sorting receptors, vesicle formation machinery, kinase-mediated regulation, organelle acidification, and, most likely, activities of the cytoskeleton.
  • Influenza attachment to the cells surface occurs via binding of the HA1 subunit to cell surface glycoproteins and glycolipids that carry oligosaccharide moieties with terminal sialic acid residues (Skehel and Wiley 2000).
  • the linkage by which the sialic acid is connected to the next saccharide contributes to species specificity.
  • Avian strains including H5N1 prefer an a-(2,3)-link and human strains a-(2,6)-link (Matrosovich 2006).
  • binding occurs preferentially to microvilli on the apical surface, and endocytosis occurs at base of these extensions (Matlin 1982).
  • Endocytic internalization occurs within a few minutes after binding (Matlin 1982; Yoshimura and Ohnishi 1984).
  • influenza virus makes use of three different types of cellular processes; 1) preexisting clathrin coated pits, 2) virus-induced clathrin coated pits, and 3) endocytosis in vesicles without visible coat (Matlin 1982; Sieczkarski and Whittaker 2002; Rust et al. 2004).
  • Video microscopy using fluorescent viruses showed the virus particles undergoing actin-mediated rapid motion in the cell periphery followed by minus end-directed, microtubule-mediated transport to the perinuclear area of the cell.
  • Live cell imaging indicated that the virus particles first entered a subpopulation of mobile, peripheral early endosomes that carry them deeper into the cytoplasm before penetration takes place (Lakadamyali et al. 2003; Rust et al. 2004).
  • the endocytic process is regulated by protein and lipid kinases, the proteasome, as well as by Rabs and ubiquitin-dependent sorting factors (Khor et al. 2003; Whittaker 2006).
  • the membrane penetration step is mediated by low pH-mediated activation of the trimeric, metastable HA, and the conversion of this Type I viral fusion protein to a membrane fusion competent conformation (Maeda et al. 1981; White et al. 1982). This occurs about 16 min after internalization, and the pH threshold varies between strains in the 5.0-5.6 range.
  • the target membrane is the limiting membrane of intermediate or late endosomes.
  • the mechanism of fusion has been extensively studied (Kielian and Rey 2006). Further it was observed that fusion itself does not seem to require any host cell components except a lipid bilayer membrane and a functional acidification system (Maeda et al. 1981; White et al. 1982).
  • the penetration step is inhibited by agents such as lysosomotropic weak bases, carboxylic ionophores, and proton pump inhibitors (Malin 1982; Whittaker 2006).
  • the capsid has to be disassembled. This step involves acidification of the viral interior through the amantadine-sensitive M2-channels causes dissociation of M1 from the vRNPs (Bukrinskaya et al. 1982; Martin and Helenius 1991; Pinto et al. 1992). Transport of the individual vRNPs to the nuclear pore complexes and transfer into the nucleus depends on cellular nuclear transport receptors (O'Neill et al. 1995; Cros et al. 2005). Replication of the viral RNAs (synthesis of positive and negative strands), and transcription occurs in complexes tightly associated with the chromatin in the nucleus.
  • RNA polymerase activating factors e.g., RNA polymerase activating factor, a chaperone HSP90, hCLE, and a human splicing factor UAP56.
  • Viral gene expression is subject to complex cellular control at the transcriptional level, a control system dependent on cellular kinases (Whittaker 2006).
  • the final assembly of an influenza particle occurs during a budding process at the plasma membrane.
  • budding occurs at the apical membrane domain only (Rodriguez-Boulan 1983).
  • the progeny vRNPs are transported within the nucleoplasm to the nuclear envelope, then from the nucleus to the cytoplasm, and finally they accumulate in the cell periphery. Exit from the nucleus is dependent on viral protein NEP and M1, and a variety of cellular proteins including CRM1 (a nuclear export receptor), caspases, and possibly some nuclear protein chaperones.
  • Phosphorylation plays a role in nuclear export by regulating M1 and NEP synthesis, and also through the MAPK/ERK system (Bui et al. 1996; Ludwig 2006).
  • G protein and protein kinase signaling is involved in influenza virus budding from infected host cells (Hui E. and Nayak D, 2002).
  • the three membrane proteins of the virus are synthesized, folded and assembled into oligomers in the ER (Doms et al. 1993). They pass through the Golgi complex; undergo maturation through modification of their carbohydrate moieties and proteolytic cleavage. After reaching the plasma membrane they associate with M1 and the vRNPs in a budding process that results in the inclusion of all eight vRNPs and exclusion of most host cell components except lipids.
  • Influenza infection is associated with activation of several signaling cascades including the MAPK pathway (ERK, JNK, p38 and BMK-1/ERK5), the IkB/NF-kB signaling module, the Raf/MEK/ERK cascade, and programmed cell death (Ludwig 2006).
  • MAPK pathway ERK, JNK, p38 and BMK-1/ERK5
  • IkB/NF-kB signaling module the Raf/MEK/ERK cascade
  • programmed cell death Lidwig 2006.
  • FIGS. 1 and 2 show a possible binding mode of the PB1 helix-derived sequence MDVNPTLLFLKVPAQ (SEQ ID NO: 1).
  • a peptidomimetic macrocycle of the invention is prepared starting with the corresponding uncrosslinked polypeptide sequence MDVNPTLLFLKVPAQ (SEQ ID NO: 1) and replacing the 7 th and 10 th amino acids with an alpha, alpha-disubstituted amino acid (e.g. the S5 olefin amino acid).
  • An olefin metathesis reaction is performed resulting in a peptidomimetic macrocycle comprising an i to i+3 crosslink as shown in FIG. 2 b.
  • Peptidomimetic macrocycles were synthesized, purified and analyzed as previously described (Walensky et al (2004) Science 305:1466-70; Walensky et al (2006) Mol Cell 24:199-210; Bernal et al (2007) J. Am Chem Soc. 9129, 2456-2457) and as indicated below.
  • the macrocycles used in this study are shown in Table 5.
  • the corresponding uncrosslinked polypeptides represent the natural counterparts of the peptidomimetic macrocycles of the invention.
  • Alpha,alpha-disubstituted non-natural amino acids containing olefinic side chains were synthesized according to Williams et al. (1991) J. Am. Chem. Soc. 113:9276; Schafmeister et al. (2000) J. Am. Chem Soc. 122:5891 and Verdine et al PCT WO 2008/121767.
  • Peptidomimetic macrocycles were designed by replacing two or more naturally occurring amino acids with the corresponding synthetic amino acids. Substitutions were made at the i and i+3, i and i+4, i and i+6, and i and i+7 positions. Macrocycles were generated by solid phase peptide synthesis followed by olefin metathesis-based crosslinking of the synthetic amino acids via their olefin-containing side chains.
  • Nle represents norleucine
  • Aib represents 2-aminoisobutyric acid
  • Ac represents acetyl
  • Pr represents propionyl.
  • Amino acids represented as “$” are alpha-Me S5-pentenyl-alanine olefin amino acids connected by an all-carbon i to i+4 crosslinker comprising one double bond.
  • Amino acids represented as “$r5” are alpha-Me R5-pentenyl-alanine olefin amino acids connected by an all-carbon i to i+4 crosslinker comprising one double bond.
  • Amino acids represented as “$s8” are alpha-Me S8-octenyl-alanine olefin amino acids connected by an all-carbon i to i+7 crosslinker comprising one double bond.
  • Amino acids represented as “$r8” are alpha-Me R8-octenyl-alanine olefin amino acids connected by an all-carbon i to i+7 crosslinker comprising one double bond.
  • “Ahx” represents an aminocyclohexyl linker.
  • the crosslinkers are linear all-carbon crosslinker comprising eight or eleven carbon atoms between the alpha carbons of each amino acid.
  • Amino acids represented as “$/” are alpha-Me S5-pentenyl-alanine olefin amino acids that are not connected by any crosslinker.
  • Amino acids represented as “$/r5” are alpha-Me R5-pentenyl-alanine olefin amino acids that are not connected by any crosslinker.
  • Amino acids represented as “$/s8” are alpha-Me S8-octenyl-alanine olefin amino acids that are not connected by any crosslinker.
  • Amino acids represented as “$/r8” are alpha-Me R8-octenyl-alanine olefin amino acids that are not connected by any crosslinker.
  • the non-natural amino acids were characterized by nuclear magnetic resonance (NMR) spectroscopy (Varian Mercury 400) and mass spectrometry (Micromass LCT). Peptide synthesis was performed either manually or on an automated peptide synthesizer (Applied Biosystems, model 433A), using solid phase conditions, rink amide AM resin (Novabiochem), and Fmoc main-chain protecting group chemistry.
  • the synthesized peptides include a norleucine replacement for methionine to avoid issues with unwanted thioether oxidation.
  • the proline residue is replaced with a 2-aminoisobutyric acid residue (Aib) to increase helicity, and the effect of Glu-to-Arg substitution on cell penetrability was also explored.
  • the N-termini of the synthetic peptides were acetylated, while the C-termini were amidated.
  • Table 5 shows a list of peptidomimetic macrocycles of the invention prepared.
  • PB1 peptidomimetic macrocycles were tested for PK properties in single N dose in rats.
  • the in-life portion of the study was conducted at ViviSource Laboratories (Waltham, Mass.).
  • a single intravenous dose of 3 mg/kg Stapled Peptide formulated in water continuing 5% PEG-400 and 2% Dextrose was administered to a pair of jugular vein-cannulated male Sprague-Dawley rats.
  • the N dose was mostly well-tolerated and animals appeared healthy within the study duration.
  • Blood samples were collected over thirteen sampling times up to 24 hours and the plasma samples were shipped on dry ice to Tandem Bioanalytical Facilities, Inc. (Woburn, Mass.) for the analytical phase of the study.
  • Quantification in plasma samples was preceded by the preparation of sample extracts by combining 50 ⁇ l of ammonium hydroxide (14.5 M ammonia), 1 mL of a 1:1 acetonitrile/methanol solution, and 50 ⁇ l of internal standard with 50 ⁇ l of each plasma sample. The mixtures were centrifuged to separate liquid supernatant from solid precipitate and supernatants were dried at 40° C. under flowing nitrogen gas. The dried sample extracts were reconstituted in 50 ⁇ l of a 1:1 water/methanol solution that contained 0.1% (v/v) trifluoroacetic acid.
  • Plasma sample extracts were analyzed by a liquid chromatography-mass spectrometry method that utilized an API 5000 (Applied Biosystems) instrument operated in positive ionization mode at a temperature of 500° C. using a multiple reaction monitoring mode of detection (MRM).
  • the analytical column for liquid chromatography was a Varian Metasil C18, 50 mm ⁇ 2 mm and mobile phases A (0.1% formic acid in water) and B (0.1% formic acid in acetonitrile) were pumped at a flow rate of 0.5 ml/min.
  • Quantification in plasma extracts was made by linear regression analysis employing a pure reference standard Stapled Peptide diluted in normal rat plasma to prepare eight calibration standards over the working concentration range of 20-10,000 ng/ml.
  • the calibration standards were extracted in identical fashion as sample extracts and analyzed before and after the sample extracts.
  • Pharmacokinetic parameters were calculated using a non-compartmental model using the PK Functions add-in for Microsoft Excel.
  • the terminal elimination half-life was calculated as ln(2)/( ⁇ z), where the rate constant ( ⁇ z) was calculated as ⁇ 1 times the estimated slope of the log-concentration versus time data over 2-12 hr.
  • AUC values (hr*ng/ml) were calculated by statistical moment and linear trapezoidal approximation methods over time points of 0-24 hours and 24 hour concentration values were divided by ( ⁇ z) was added in order to extrapolate AUMC and AUC values to infinite time.
  • Total body clearance (per kg body weight) was calculated as dose divided by AUC.
  • Vss volume of distribution at steady state
  • MRT mean residence time

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pulmonology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Novel peptidomimetic macrocycles and methods of using such macrocycles for the treatment of viral diseases are described.

Description

CROSS REFERENCE
This application is a continuation application of Ser. No. 14/853,894, filed Sep. 14, 2015, which is a continuation of U.S. application Ser. No. 13/129,118, filed Nov. 1, 2011, now U.S. Pat. No. 9,175,047, which is a U.S. National Phase Application under 35 U.S.C. § 371 of PCT/US2010/021091, filed Jan. 14, 2010, which claims the benefit of U.S. Provisional Application No. 61/144,706, filed Jan. 14, 2009, the content of each of which is incorporated herein by reference in its entirety.
SEQUENCE LISTING
The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Apr. 7, 2016, is named 35224-724.303_SL.txt and is 82,255 bytes in size.
BACKGROUND OF THE INVENTION
Seasonal influenza infection is a major health concern for first-world and developing nations alike. Each year in the United States, five- to twenty-percent of the population gets the flu, more than 200,000 people are hospitalized from flu complications, and about 36,000 people die from flu. Worldwide, influenza causes tens of millions of respiratory illnesses and 250,000 to 500,000 deaths each year. New strains of avian influenza that are transmissible to humans are a critical concern for global health because these flu strains could yield pandemic disease for which no immunity exists, potentially resulting in millions of fatalities. “Avian flu” refers to a pathogenic avian influenza subtype that is highly contagious among birds and causes high mortality among domestic poultry. Outbreaks of avian flu among poultry and wild birds are ongoing in a number of countries, and at least three subgroups of avian flu viruses have infected humans to date. While avian flu infections of humans are rare, and most cases have been associated with direct poultry contact during outbreaks among livestock, infection in humans is very serious when it does occur: to date, over half of all reported human cases have been fatal. Since first reported in Hong Kong in 1996, the World Health Organization has carefully tracked avian flu and instances of animal-to-human influenza transmission, with confirmed cases reported from China, Indonesia, and Southeast Asia; Pakistan; Iraq; Egypt; and elsewhere, with 385 cases resulting in 243 deaths worldwide. While there is no evidence of sustained human-to-human transmission, instances of human-to-human spread of avian flu may have occurred. Since all influenza viruses have the ability to rapidly mutate, there is considerable concern that avian flu may be able to infect humans more easily and become communicable from one person to another. Also, avian flu virus strains have not infected many humans worldwide, so there is little or no immune protection against these strains in the human population; therefore, an influenza pandemic could easily occur if sustained avian flu virus transmission were to develop.
Three classes of influenza viruses, A, B and C, are responsible for human flu, with influenza A and B viruses causing seasonal epidemics of disease almost every winter. Influenza A viruses are divided into subtypes based on characteristics of two proteins, hemagglutinin (H) and neuraminidase (N), on the surface of the virus. There are 16 different hemagglutinin subtypes and 9 different neuraminidase subtypes, with H1N1 and H3N2 being the most common subtypes found in humans. The avian flu virus refers to influenza A H5N1. Influenza A is a negative-sense (3′ to 5′) single-stranded RNA virus. Its viral genome, which encodes 11 proteins (HA, NA, NP, M1, M2, NS1, NEP, PA, PB1, PB1-F2, PB2) in its RNA, cannot be translated into protein directly; rather, the virus depends on its RNA-dependent RNA polymerase to transcribe its genome to positive-sense RNA prior to translation. RNA-dependent RNA polymerases have no mammalian counterpart, which renders species selectivity less problematic in the development of therapeutics that target this enzyme. Other examples of viral RNA-dependent RNA polymerases include polioviral 3Dpol, vesicular stomatitis virus L, and hepatitis C virus NS5b; the latter is an active target for development of hepatitis C antiviral therapies. Unlike current flu targets (e.g., neuraminidase for Tamiflu), the influenza RNA polymerase is highly conserved and therefore less likely to suffer the resistance issues that current drugs face.
Recently, researchers reported the first atomic-resolution structural details of the influenza protein RNA polymerase, a critical enzyme for viral replication and a novel target for both therapeutic intervention and prophylaxis during influenza outbreaks (He, X., et al., Nature, 2008. 454: p. 1123-6; Obayashi, E., et al., Nature, 2008. 454: p. 1127-31). The influenza RNA-dependent RNA polymerase is a heterotrimer of three subunits, PA, PB1, and PB2, with the 310-helical N-terminal region of PB1 binding between the “jaws” of the PA protein. The PB1 helix is thought to be important for complex formation and nuclear transport and inhibits influenza A viral replication by interfering with polymerase activity. Recently, the PB2 subunit has also been shown to play an essential role in activity of the viral polymerase complex, for instance through contacts with the PB1 subunit. See Sugiyama et al, EMBO Journal, 2009, 28, 1803-1811. However, little is known about compounds capable of interfering with the binding and activity of these proteins. In general, there remains a need for therapeutic methods of treating viral diseases in which RNA-dependent RNA polymerases play a role, and for compositions and methods capable of modifying the activity such polymerases.
SUMMARY OF THE INVENTION
The present invention addresses these and other needs. In one aspect, the present invention provides a peptidomimetic macrocycle capable of binding to a viral RNA-dependent RNA polymerase. Such a macrocycle may, for example, be capable of disrupting the assembly of subunits of a viral RNA-dependent RNA polymerase complex. In one embodiment, such a macrocycle may compete with the binding of a peptide of the sequence MDVNPTLLFLKVPAQ (SEQ ID NO: 1) or MERIKELRNLM (SEQ ID NO: 2) to said viral RNA-dependent RNA polymerase. In one embodiment, a peptidomimetic macrocycle of the invention comprises an amino acid sequence which is at least about 60%, 80%, 90%, or 95% identical to the amino acid sequence MDVNPTLLFLKVPAQ (PB1) (SEQ ID NO: 1) or MERIKELRNLM (PB2) (SEQ ID NO: 2). Alternatively, the amino acid sequence of said peptidomimetic macrocycle is identified and optimized for its ability to bind to either the PB1 peptide binding site of the PA protein or the PB2 peptide binding site of the PB1 protein, for example through affinity selection with the PA or PB1 target protein or by structure-based design, with such a mechanism of action being confirmed by biophysical/structural studies and/or competitive displacement assays with the PB1 or PB2 peptide. In some embodiments, the peptidomimetic macrocycle comprises a helix, such as a 310 helix or an α-helix. In other embodiments, the peptidomimetic macrocycle comprises an α,α-disubstituted amino acid. A peptidomimetic macrocycle of the invention may comprise a crosslinker linking the α-positions of at least two amino acids. At least one of said two amino acids may be an α,α-disubstituted amino acid.
In some embodiments, the peptidomimetic macrocycle has the formula:
Figure US10022422-20180717-C00001
wherein:
each A, C, D, and E is independently a natural or non-natural amino acid;
B is a natural or non-natural amino acid, amino acid analog,
Figure US10022422-20180717-C00002

[—NH-L3-CO—], [—NH-L3-SO2—], or [—NH-L3-];
R1 and R2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-;
R3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5;
L is a macrocycle-forming linker of the formula -L1-L2-;
L1 and L2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R4—K—R4—]n, each being optionally substituted with R5;
each R4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
each K is O, S, SO, SO2, CO, CO2, or CONR3;
each R5 is independently halogen, alkyl, —OR6, —N(R6)2, —SR6, —SOR6, —SO2R6, —CO2R6, a fluorescent moiety, a radioisotope or a therapeutic agent;
each R6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
R7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5, or part of a cyclic structure with a D residue;
R8 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5, or part of a cyclic structure with an E residue;
v and w are independently integers from 1-1000;
u, x, y and z are independently integers from 0-10; and
n is an integer from 1-5.
In other embodiments, the peptidomimetic macrocycle may comprise a crosslinker linking a backbone amino group of a first amino acid to a second amino acid within the peptidomimetic macrocycle. For example, the invention provides peptidomimetic macrocycles of the formula (IV) or (IVa):
Figure US10022422-20180717-C00003
wherein:
each A, C, D, and E is independently a natural or non-natural amino acid;
B is a natural or non-natural amino acid, amino acid analog,
Figure US10022422-20180717-C00004

[—NH-L3-CO—], [—NH-L3-SO2—], or [—NH-L3-];
R1 and R2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-, or part of a cyclic structure with an E residue;
R3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5;
L1 and L2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R4—K—R4—]n, each being optionally substituted with R5;
each R4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
each K is O, S, SO, SO2, CO, CO2, or CONR3;
each R5 is independently halogen, alkyl, —OR6, —N(R6)2, —SR6, —SOR6, —SO2R6, —CO2R6, a fluorescent moiety, a radioisotope or a therapeutic agent;
each R6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
R7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5;
v and w are independently integers from 1-1000;
u, x, y and z are independently integers from 0-10; and
n is an integer from 1-5.
In some embodiments, x+y+z is 2, 3, 5 or 6.
Additionally, the invention provides a method of treating influenza virus infection in a subject comprising administering to the subject a peptidomimetic macrocycle of the invention. Also provided is a method of preventing infection by an influenza virus in a subject comprising administering to the subject a peptidomimetic macrocycle of the invention, or a method of inhibiting the activity of the RNA-dependent RNA polymerase of an influenza virus in a subject comprising administering to the subject such a peptidomimetic macrocycle.
INCORPORATION BY REFERENCE
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
BRIEF DESCRIPTION OF THE DRAWINGS
The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
FIG. 1a shows a bound PB1 helix in complex with the PA subunit of a RNA-dependent RNA polymerase. Leu7 and Leu10 (light color) are candidate residues for i, i+3 macrocycle formation to stabilize a 310 helix.
FIG. 1b shows a macrocycle derived from the sequence in FIG. 1 a.
FIG. 2a shows the sequence of FIG. 1a excised from its complex with the PA subunit or a RNA-dependent RNA polymerase.
FIG. 2b shows the macrocycle derived from the sequence in FIG. 1b excised from its complex with the PA subunit of a RNA-dependent RNA polymerase.
FIG. 3 describes the plasma stability of several peptidomimetic macrocycles of the invention.
FIG. 4 shows in vivo pharmacokinetic properties of several peptidomimetic macrocycles of the invention.
FIGS. 5a-5f illustrate in vivo pharmacokinetic properties of peptidomimetic macrocycles of the invention.
FIG. 6 shows selected pharmacokinetic parameters for several peptidomimetic macrocycles of the invention.
FIG. 7 illustrates pharmacokinetic properties for intravenous and subcutaneous modes of administration for a peptidomimetic macrocycle of the invention.
DETAILED DESCRIPTION OF THE INVENTION
As used herein, the term “macrocycle” refers to a molecule having a chemical structure including a ring or cycle formed by at least 9 covalently bonded atoms.
As used herein, the term “peptidomimetic macrocycle” or “crosslinked polypeptide” refers to a compound comprising a plurality of amino acid residues joined by a plurality of peptide bonds and at least one macrocycle-forming linker which forms a macrocycle between a first naturally-occurring or non-naturally-occurring amino acid residue (or analog) and a second naturally-occurring or non-naturally-occurring amino acid residue (or analog) within the same molecule. Peptidomimetic macrocycle include embodiments where the macrocycle-forming linker connects the α carbon of the first amino acid residue (or analog) to the α carbon of the second amino acid residue (or analog). The peptidomimetic macrocycles optionally include one or more non-peptide bonds between one or more amino acid residues and/or amino acid analog residues, and optionally include one or more non-naturally-occurring amino acid residues or amino acid analog residues in addition to any which form the macrocycle. A “corresponding uncrosslinked polypeptide” when referred to in the context of a peptidomimetic macrocycle is understood to relate to a polypeptide of the same length as the macrocycle and comprising the equivalent natural amino acids of the wild-type sequence corresponding to the macrocycle.
As used herein, the term “stability” refers to the maintenance of a defined secondary structure in solution by a peptidomimetic macrocycle of the invention as measured by circular dichroism, NMR or another biophysical measure, or resistance to proteolytic degradation in vitro or in vivo. Non-limiting examples of secondary structures contemplated in this invention are helices, β-turns, and β-pleated sheets. In general, the term “helix” or “helical” is used to refer to any type of helical secondary structure, including 310-helices, α-helices, and π-helices.
As used herein, the term “helical stability” refers to the maintenance of helical structure by a peptidomimetic macrocycle of the invention as measured by circular dichroism or NMR. For example, in some embodiments, the peptidomimetic macrocycles of the invention exhibit at least a 1.25, 1.5, 1.75 or 2-fold increase in helicity as determined by circular dichroism compared to a corresponding uncrosslinked macrocycle.
The term “α-amino acid” or simply “amino acid” refers to a molecule containing both an amino group and a carboxyl group bound to a carbon which is designated the α-carbon. Suitable amino acids include, without limitation, both the D- and L-isomers of the naturally-occurring amino acids, as well as non-naturally occurring amino acids prepared by organic synthesis or other metabolic routes. Unless the context specifically indicates otherwise, the term amino acid, as used herein, is intended to include amino acid analogs.
The term “naturally occurring amino acid” refers to any one of the twenty amino acids commonly found in peptides synthesized in nature, and known by the one letter abbreviations A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y and V.
The term “amino acid analog” or “non-natural amino acid” refers to a molecule which is structurally similar to an amino acid and which can be substituted for an amino acid in the formation of a peptidomimetic macrocycle. Amino acid analogs include, without limitation, compounds which are structurally identical to an amino acid, as defined herein, except for the inclusion of one or more additional methylene groups between the amino and carboxyl group (e.g., α-amino β-carboxy acids), or for the substitution of the amino or carboxy group by a similarly reactive group (e.g., substitution of the primary amine with a secondary or tertiary amine, or substitution or the carboxy group with an ester).
A “non-essential” amino acid residue is a residue that can be altered from the wild-type sequence of a polypeptide (e.g., a BH3 domain or the p53 MDM2 binding domain) without abolishing or substantially altering its essential biological or biochemical activity (e.g., receptor binding or activation). An “essential” amino acid residue is a residue that, when altered from the wild-type sequence of the polypeptide, results in abolishing or substantially abolishing the polypeptide's essential biological or biochemical activity.
A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., K, R, H), acidic side chains (e.g., D, E), uncharged polar side chains (e.g., G, N, Q, S, T, Y, C), nonpolar side chains (e.g., A, V, L, I, P, F, M, W), beta-branched side chains (e.g., T, V, I) and aromatic side chains (e.g., Y, F, W, H). Thus, a predicted nonessential amino acid residue in a polypeptide, for example, is preferably replaced with another amino acid residue from the same side chain family. Other examples of acceptable substitutions are substitutions based on isosteric considerations (e.g. norleucine for methionine) or other properties (e.g. 2-thienylalanine for phenylalanine).
The term “member” as used herein in conjunction with macrocycles or macrocycle-forming linkers refers to the atoms that form or can form the macrocycle, and excludes substituent or side chain atoms. By analogy, cyclodecane, 1,2-difluoro-decane and 1,3-dimethyl cyclodecane are all considered ten-membered macrocycles as the hydrogen or fluoro substituents or methyl side chains do not participate in forming the macrocycle.
The symbol “
Figure US10022422-20180717-P00001
” when used as part of a molecular structure refers to a single bond or a trans or cis double bond.
The term “amino acid side chain” refers to a moiety attached to the α-carbon in an amino acid. For example, the amino acid side chain for alanine is methyl, the amino acid side chain for phenylalanine is phenylmethyl, the amino acid side chain for cysteine is thiomethyl, the amino acid side chain for aspartate is carboxymethyl, the amino acid side chain for tyrosine is 4-hydroxyphenylmethyl, etc. Other non-naturally occurring amino acid side chains are also included, for example, those that occur in nature (e.g., an amino acid metabolite) or those that are made synthetically (e.g., an α,α di-substituted amino acid).
The term “α,α di-substituted amino” acid refers to a molecule or moiety containing both an amino group and a carboxyl group bound to a carbon (the α-carbon) that is attached to two natural or non-natural amino acid side chains.
The term “polypeptide” encompasses two or more naturally or non-naturally-occurring amino acids joined by a covalent bond (e.g., an amide bond). Polypeptides as described herein include full length proteins (e.g., fully processed proteins) as well as shorter amino acid sequences (e.g., fragments of naturally-occurring proteins or synthetic polypeptide fragments).
The term “macrocyclization reagent” or “macrocycle-forming reagent” as used herein refers to any reagent which may be used to prepare a peptidomimetic macrocycle of the invention by mediating the reaction between two reactive groups. Reactive groups may be, for example, an azide and alkyne, in which case macrocyclization reagents include, without limitation, Cu reagents such as reagents which provide a reactive Cu(I) species, such as CuBr, CuI or CuOTf, as well as Cu(II) salts such as Cu(CO2CH3)2, CuSO4, and CuCl2 that can be converted in situ to an active Cu(I) reagent by the addition of a reducing agent such as ascorbic acid or sodium ascorbate. Macrocyclization reagents may additionally include, for example, Ru reagents known in the art such as Cp*RuCl(PPh3)2, [Cp*RuCl]4 or other Ru reagents which may provide a reactive Ru(II) species. In other cases, the reactive groups are terminal olefins. In such embodiments, the macrocyclization reagents or macrocycle-forming reagents are metathesis catalysts including, but not limited to, stabilized, late transition metal carbene complex catalysts such as Group VIII transition metal carbene catalysts. For example, such catalysts are Ru and Os metal centers having a +2 oxidation state, an electron count of 16 and pentacoordinated. Additional catalysts are disclosed in Grubbs et al., “Ring Closing Metathesis and Related Processes in Organic Synthesis” Acc. Chem. Res. 1995, 28, 446452, and U.S. Pat. No. 5,811,515. In yet other cases, the reactive groups are thiol groups. In such embodiments, the macrocyclization reagent is, for example, a linker functionalized with two thiol-reactive groups such as halogen groups.
The term “halo” or “halogen” refers to fluorine, chlorine, bromine or iodine or a radical thereof.
The term “alkyl” refers to a hydrocarbon chain that is a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C1-C10 indicates that the group has from 1 to 10 (inclusive) carbon atoms in it. In the absence of any numerical designation, “alkyl” is a chain (straight or branched) having 1 to 20 (inclusive) carbon atoms in it.
The term “alkylene” refers to a divalent alkyl (i.e., —R—).
The term “alkenyl” refers to a hydrocarbon chain that is a straight chain or branched chain having one or more carbon-carbon double bonds. The alkenyl moiety contains the indicated number of carbon atoms. For example, C2-C10 indicates that the group has from 2 to 10 (inclusive) carbon atoms in it. The term “lower alkenyl” refers to a C2-C6 alkenyl chain. In the absence of any numerical designation, “alkenyl” is a chain (straight or branched) having 2 to 20 (inclusive) carbon atoms in it.
The term “alkynyl” refers to a hydrocarbon chain that is a straight chain or branched chain having one or more carbon-carbon triple bonds. The alkynyl moiety contains the indicated number of carbon atoms. For example, C2-C10 indicates that the group has from 2 to 10 (inclusive) carbon atoms in it. The term “lower alkynyl” refers to a C2-C6 alkynyl chain. In the absence of any numerical designation, “alkynyl” is a chain (straight or branched) having 2 to 20 (inclusive) carbon atoms in it.
The term “aryl” refers to a 6-carbon monocyclic or 10-carbon bicyclic aromatic ring system wherein 0, 1, 2, 3, or 4 atoms of each ring are substituted by a substituent. Examples of aryl groups include phenyl, naphthyl and the like.
The term “arylalkyl” or the term “aralkyl” refers to alkyl substituted with an aryl. The term “arylalkoxy” refers to an alkoxy substituted with aryl.
“Arylalkyl” refers to an aryl group, as defined above, wherein one of the aryl group's hydrogen atoms has been replaced with a C1-C5 alkyl group, as defined above. Representative examples of an arylalkyl group include, but are not limited to, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 2-propylphenyl, 3-propylphenyl, 4-propylphenyl, 2-butylphenyl, 3-butylphenyl, 4-butylphenyl, 2-pentylphenyl, 3-pentylphenyl, 4-pentylphenyl, 2-isopropylphenyl, 3-isopropylphenyl, 4-isopropylphenyl, 2-isobutylphenyl, 3-isobutylphenyl, 4-isobutylphenyl, 2-sec-butylphenyl, 3-sec-butylphenyl, 4-sec-butylphenyl, 2-t-butylphenyl, 3-t-butylphenyl and 4-t-butylphenyl.
“Arylamido” refers to an aryl group, as defined above, wherein one of the aryl group's hydrogen atoms has been replaced with one or more —C(O)NH2 groups. Representative examples of an arylamido group include 2-C(O)NH2-phenyl, 3-C(O)NH2-phenyl, 4-C(O)NH2-phenyl, 2-C(O)NH2-pyridyl, 3-C(O)NH2-pyridyl, and 4-C(O)NH2-pyridyl,
“Alkylheterocycle” refers to a C1-C5 alkyl group, as defined above, wherein one of the C1-C5 alkyl group's hydrogen atoms has been replaced with a heterocycle. Representative examples of an alkylheterocycle group include, but are not limited to, —CH2CH2-morpholine, —CH2CH2-piperidine, —CH2CH2CH2-morpholine, and —CH2CH2CH2-imidazole.
“Alkylamido” refers to a C1-C5 alkyl group, as defined above, wherein one of the C1-C5 alkyl group's hydrogen atoms has been replaced with a —C(O)NH2 group. Representative examples of an alkylamido group include, but are not limited to, —CH2—C(O)NH2, —CH2CH2—C(O)NH2, —CH2CH2CH2C(O)NH2, —CH2CH2CH2CH2C(O)NH2, —CH2CH2CH2CH2CH2C(O)NH2, —CH2CH(C(O)NH2)CH3, —CH2CH(C(O)NH2)CH2CH3, —CH(C(O)NH2)CH2CH3, —C(CH3)2CH2C(O)NH2, —CH2—CH2—NH—C(O)—CH3, —CH2—CH2—NH—C(O)—CH3—CH3, and —CH2—CH2—NH—C(O)—CH═CH2.
“Alkanol” refers to a C1-C5 alkyl group, as defined above, wherein one of the C1-C5 alkyl group's hydrogen atoms has been replaced with a hydroxyl group. Representative examples of an alkanol group include, but are not limited to, —CH2OH, —CH2CH2OH, —CH2CH2CH2OH, —CH2CH2CH2CH2OH, —CH2CH2CH2 CH2CH2OH, —CH2CH(OH)CH3, —CH2CH(OH)CH2CH3, —CH(OH)CH3 and —C(CH3)2CH2OH.
“Alkylcarboxy” refers to a C1-C5 alkyl group, as defined above, wherein one of the C1-C5 alkyl group's hydrogen atoms has been replaced with a —COOH group. Representative examples of an alkylcarboxy group include, but are not limited to, —CH2COOH, —CH2CH2COOH, —CH2CH2CH2COOH, —CH2CH2CH2CH2COOH, —CH2CH(COOH)CH3, —CH2CH2CH2CH2CH2COOH, —CH2CH(COOH)CH2CH3, —CH(COOH)CH2CH3 and —C(CH3)2CH2COOH.
The term “cycloalkyl” as employed herein includes saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12 carbons, preferably 3 to 8 carbons, and more preferably 3 to 6 carbons, wherein the cycloalkyl group additionally is optionally substituted. Some cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl.
The term “heteroaryl” refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of O, N, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2, 3, or 4 atoms of each ring are substituted by a substituent. Examples of heteroaryl groups include pyridyl, furyl or furanyl, imidazolyl, benzimidazolyl, pyrimidinyl, thiophenyl or thienyl, quinolinyl, indolyl, thiazolyl, and the like.
The term “heteroarylalkyl” or the term “heteroaralkyl” refers to an alkyl substituted with a heteroaryl. The term “heteroarylalkoxy” refers to an alkoxy substituted with heteroaryl.
The term “heteroarylalkyl” or the term “heteroaralkyl” refers to an alkyl substituted with a heteroaryl. The term “heteroarylalkoxy” refers to an alkoxy substituted with heteroaryl.
The term “heterocyclyl” refers to a nonaromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of O, N, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring are substituted by a substituent. Examples of heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and the like.
The term “substituent” refers to a group replacing a second atom or group such as a hydrogen atom on any molecule, compound or moiety. Suitable substituents include, without limitation, halo, hydroxy, mercapto, oxo, nitro, haloalkyl, alkyl, alkaryl, aryl, aralkyl, alkoxy, thioalkoxy, aryloxy, amino, alkoxycarbonyl, amido, carboxy, alkanesulfonyl, alkylcarbonyl, and cyano groups.
In some embodiments, the compounds of this invention contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of these compounds are included in the present invention unless expressly provided otherwise. In some embodiments, the compounds of this invention are also represented in multiple tautomeric forms, in such instances, the invention includes all tautomeric forms of the compounds described herein (e.g., if alkylation of a ring system results in alkylation at multiple sites, the invention includes all such reaction products). All such isomeric forms of such compounds are included in the present invention unless expressly provided otherwise. All crystal forms of the compounds described herein are included in the present invention unless expressly provided otherwise.
As used herein, the terms “increase” and “decrease” mean, respectively, to cause a statistically significantly (i.e., p<0.1) increase or decrease of at least 5%.
As used herein, the recitation of a numerical range for a variable is intended to convey that the invention may be practiced with the variable equal to any of the values within that range. Thus, for a variable which is inherently discrete, the variable is equal to any integer value within the numerical range, including the end-points of the range. Similarly, for a variable which is inherently continuous, the variable is equal to any real value within the numerical range, including the end-points of the range. As an example, and without limitation, a variable which is described as having values between 0 and 2 takes the values 0, 1 or 2 if the variable is inherently discrete, and takes the values 0.0, 0.1, 0.01, 0.001, or any other real values ≥0 and ≤2 if the variable is inherently continuous.
As used herein, unless specifically indicated otherwise, the word “or” is used in the inclusive sense of “and/or” and not the exclusive sense of “either/or.”
The term “on average” represents the mean value derived from performing at least three independent replicates for each data point.
The term “biological activity” encompasses structural and functional properties of a macrocycle of the invention. Biological activity is, for example, structural stability, helicity (including, e.g. alpha-helicity), affinity for a target, resistance to proteolytic degradation, cell penetrability, intracellular stability, in vivo stability, or any combination thereof.
The details of one or more particular embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
Design of Peptidomimetic Macrocycles
In general, peptidomimetic macrocycles are prepared that target or interact with proteins that a virus needs for infection or replication within a host cell. Such viruses may be, for example, influenza viruses belonging to Orthomyxoviridae family of viruses. This family also includes Thogoto viruses and Dhoriviruses. There are several types and subtypes of influenza viruses known, which infect humans and other species. Influenza type A viruses infect people, birds, pigs, horses, seals and other animals, but wild birds are the natural hosts for these viruses. Influenza type A viruses are divided into subtypes and named on the basis of two proteins on the surface of the virus: hemagglutinin (HA) and neuraminidase (NA). For example, an “H7N2 virus” designates an influenza A subtype that has an HA 7 protein and an NA 2 protein. Similarly an “H5N1” virus has an HA 5 protein and an NA 1 protein. There are 16 known HA subtypes and 9 known NA subtypes. Many different combinations of HA and NA proteins are possible. Only some influenza A subtypes (i.e., H1N1, H1N2, and H3N2) are currently in general circulation among people. Other subtypes are found most commonly in other animal species. For example, H7N7 and H3N8 viruses cause illness in horses, and H3N8 also has recently been shown to cause illness in dogs.
Antiviral agents according to the invention can be used to protect high-risk groups (hospital units, institutes caring for elderly, immuno-suppressed individuals), and on a case by case basis. A potential use for antiviral agents is to limit the spread and severity of the future pandemics whether caused by avian H5N1 or other strains of influenza virus. Avian influenza A viruses of the subtypes H5 and H7, including H5N1, H7N7, and H7N3 viruses, have been associated with high pathogenicity, and human infection with these viruses have ranged from mild (H7N3, H7N7) to severe and fatal disease (H7N7, H5N1). Human illness due to infection with low pathogenicity viruses has been documented, including very mild symptoms (e.g., conjunctivitis) to influenza-like illness. Examples of low pathogenicity viruses that have infected humans include H7N7, H9N2, and H7N2.
Influenza B viruses are usually found in humans but can also infect seals. Unlike influenza A viruses, these viruses are not classified according to subtype. Influenza B viruses can cause morbidity and mortality among humans, but in general are associated with less severe epidemics than influenza A viruses. Although influenza type B viruses can cause human epidemics, they have not caused pandemics.
Influenza type C viruses cause mild illness in humans and do not cause epidemics or pandemics. These viruses can also infect dogs and pigs. These viruses are not classified according to subtype.
Influenza viruses differ from each other in respect to cell surface receptor specificity and cell tropism, however they use common entry pathways. Charting these pathways and identification of host cell proteins involved in virus influenza transmission, entry, replication, biosynthesis, assembly, or exit allows the development of general agents against existing and emerging strains of influenza. The agents may also prove useful against unrelated viruses that use similar pathways. For example, the agents may protect airway epithelial cells against a number of different viruses in addition to influenza viruses.
In one embodiment the targeted virus is an adenovirus. Adenoviruses most commonly cause respiratory illness; symptoms of respiratory illness caused by adenovirus infection range from the common cold syndrome to pneumonia, croup, and bronchitis. Patients with compromised immune systems are especially susceptible to severe complications of adenovirus infection. Acute respiratory disease (ARD), first recognized among military recruits during World War II, can be caused by adenovirus infections during conditions of crowding and stress. Adenoviruses are medium-sized (90-100 nm), nonenveloped icosohedral viruses containing double-stranded DNA. There are 49 immunologically distinct types (6 subgenera: A through F) that can cause human infections. Adenoviruses are unusually stable to chemical or physical agents and adverse pH conditions, allowing for prolonged survival outside of the body. Some adenoviruses, such as AD2 and Ad5 (species C) use clathrin mediated endocytosis and macropinocytosis for infectious entry. Other adenoviruses, such as Ad3 (species B) use dynamin dependent endocytosis and macropinocytosis for infectious entry.
In one embodiment the targeted virus is a respiratory syncytial virus (RSV). RSV is the most common cause of bronchiolitis and pneumonia among infants and children under 1 year of age. Illness begins most frequently with fever, runny nose, cough, and sometimes wheezing. During their first RSV infection, between 25% and 40% of infants and young children have signs or symptoms of bronchiolitis or pneumonia, and 0.5% to 2% require hospitalization. Most children recover from illness in 8 to 15 days. The majority of children hospitalized for RSV infection are under 6 months of age. RSV also causes repeated infections throughout life, usually associated with moderate-to-severe cold-like symptoms; however, severe lower respiratory tract disease may occur at any age, especially among the elderly or among those with compromised cardiac, pulmonary, or immune systems. RSV is a negative-sense, enveloped RNA virus. The virion is variable in shape and size (average diameter of between 120 and 300 nm), is unstable in the environment (surviving only a few hours on environmental surfaces), and is readily inactivated with soap and water and disinfectants.
In one embodiment the targeted virus is a human parainfluenza virus (HPIV). HPIVs arc second to respiratory syncytial virus (RSV) as a common cause of lower respiratory tract disease in young children. Similar to RSV, HPIVs can cause repeated infections throughout life, usually manifested by an upper respiratory tract illness (e.g., a cold and/or sore throat). HPIVs can also cause serious lower respiratory tract disease with repeat infection (e.g., pneumonia, bronchitis, and bronchiolitis), especially among the elderly, and among patients with compromised immune systems. Each of the four HPIVs has different clinical and epidemiologic features. The most distinctive clinical feature of HPIV-1 and HPIV-2 is croup (i.e., laryngotracheobronchitis); HPIV-1 is the leading cause of croup in children, whereas HPIV-2 is less frequently detected. Both HPIV-1 and -2 can cause other upper and lower respiratory tract illnesses. HPIV-3 is more often associated with bronchiolitis and pneumonia. HPIV-4 is infrequently detected, possibly because it is less likely to cause severe disease. The incubation period for HPIVs is generally from 1 to 7 days. HPIVs are negative-sense, single-stranded RNA viruses that possess fusion and hemagglutinin-neuraminidase glycoprotein “spikes” on their surface. There are four serotypes types of HPIV (1 through 4) and two subtypes (4a and 4h). The virion varies in size (average diameter between 150 and 300 nm) and shape, is unstable in the environment (surviving a few hours on environmental surfaces), and is readily inactivated with soap and water.
In one embodiment the targeted virus is a coronavirus. Coronavirus is a genus of animal virus belonging to the family Coronaviridae. Coronaviruses are enveloped viruses with a positive-sense single-stranded RNA genome and a helical symmetry. The genomic size of coronaviruses ranges from approximately 16 to 31 kilobases, extraordinarily large for an RNA virus. The name “coronavirus” is derived from the Latin corona, meaning crown, as the virus envelope appears under electron microscopy to be crowned by a characteristic ring of small bulbous structures. This morphology is actually formed by the viral spike peplomers, which are proteins that populate the surface of the virus and determine host tropism. Coronaviruses are grouped in the order Nidovirales, named for the Latin nidus, meaning nest, as all viruses in this order produce a 3′ co-terminal nested set of subgenomic mRNA's during infection. Proteins that contribute to the overall structure of all coronaviruses are the spike, envelope, membrane and nucleocapsid. In the specific case of SARS a defined receptor-binding domain on S mediates the attachment of the virus to its cellular receptor, angiotensin-converting enzyme 2.
In one embodiment the targeted virus is a rhinovirus. Rhinovirus (from the Greek rhin-, which means “nose”) is a genus of the Picornaviridae family of viruses. Rhinoviruses are the most common viral infective agents in humans, and a causative agent of the common cold. There are over 105 serologic virus types that cause cold symptoms, and rhinoviruses are responsible for approximately 50% of all cases. Rhinoviruses have single-stranded positive sense RNA genomes of between 7.2 and 8.5 kb in length. At the 5′ end of the genome is a virus-encoded protein, and like mammalian mRNA, there is a 3′ poly-A tail. Structural proteins are encoded in the 5′ region of the genome and non structural at the end. This is the same for all picornaviruses. The viral particles themselves are not enveloped and are icosahedral in structure.
Any secondary structure of a viral protein (or of a host cell protein involved in viral infectivity) can form the basis of the methods of the invention. For example, a viral proteins comprising a secondary structure which is a helix may be used to design peptidomimetic macrocycles based on said helix.
In one embodiment, the peptidomimetic macrocycle of the invention is designed based on the PB1 or PB2 sequence of an influenza virus. The PB1 sequence is highly conserved across all known strains of influenza A virus, which may result in less drug resistance should than that observed with the current standard of care. An alignment of the first 25 N-terminal amino acids of PB1 from the NCBI data bank's 2,485 influenza A virus strains (Ghanem, 2007) demonstrates the remarkable sequence conservation in the PA interaction domain of PB1. Therefore, antiviral therapies based on the PB1 sequence may block most if not all, influenza A virus strains. Additionally, sequence modification of a peptidomimetic macrocycle based on these few variations in PB1 may enable an antiviral cocktail of PB1 inhibitors to eliminate resistance due to escape mutants.
A non-limiting exemplary list of sequences suitable for macrocyclization as well as macrocyclic peptides for use in the present invention is given below:
TABLE 1a
Exemplary PB1 peptidomimetic macrocycles of the invention.
Influenza PB1 Sequences For Macrocyclization (bold = critical residue; X = cross-linked amino acid) SEQ ID
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 NO:
Ac— Met Asp Val Asn Pro Thr Leu Leu Phe Leu Lys Val Pro Ala Gln —NH2 3 wt,
backbone
H— bonds
Ac— Met Asp Val Asn Pro Thr Leu Leu Phe Leu Lys Val Pro Ala Gln —NH2 4 wt,
side-chain
H— bonds
contacts
Ac— Met Asp Val Asn Pro Thr Leu Leu Phe Leu Lys Val Pro Ala Gln —NH2 5 wt,
side-chain
hydrophobic
contacts
Ac— Met/ Asp Val Asn X Thr/ Leu X Phe Leu Lys Val Pro/ Ala/ Gln/ —NH2 6 i--> i + 3
Nle Aib Aib Aib Arg x-link #1
Ac— Met/ Asp Val Asn Pro/ X Leu Leu X Leu Lys Val Pro/ Ala/ Gln/ —NH2 7 i--> i + 3
Nle Aib Aib Aib Arg x-link #2
Ac— Met/ Asp Val Asn Pro/ Thr/ X Leu Phe X Lys Val Pro/ Ala/ Gln/ —NH2 8 i--> i + 3
Nle Aib Aib Aib Aib Arg x-link #3
Ac— Met/ Asp Val Asn Pro/ Thr/ Leu Leu X Leu Lys X Pro/ Ala/ Gln/ —NH2 9 i--> i + 3
Nle Aib Aib Aib Aib Arg x-link #4
Ac— Met/ Asp Val Asn Pro/ Thr/ Leu Leu Phe X Lys Val X Ala/ Gln/ —NH2 10 i--> i + 3
Nle Aib Aib Aib Arg x-link #5
Ac— Met/ Asp Val Asn Pro/ Thr/ Leu Leu Phe Leu Lys X Pro/ Ala/ X —NH2 11 i --> i + 3
Nle Aib Aib Aib Aib x-link #6
X Thr/ Leu Leu Phe Leu Lys Val Pro/ Ala/ Gln/ —NH2 12 Formula IV
Aib Aib Aib Arg x-link #1
PB1 Peptidomimetic Macrocycles (bold = mutated residue; $ = S5-olefin amino acid; % = Formula IV x-link) SEQ ID Charge at
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 NO: pH 7.4
Ac— Met Asp Val Asn $ Thr Leu $ Phe Leu Lys Val Pro Ala Gln —NH2 13 0
Ac— Nle Asp Val Asn $ Aib Leu $ Phe Leu Lys Val Aib Aib Gln —NH2 14 0
Ac— Nle Asp Val Asn $ Aib Leu $ Phe Leu Lys Val Aib Aib Arg —NH2 15 1
Ac— Met Asp Val Asn Pro $ Leu Leu $ Leu Lys Val Pro Ala Gln —NH2 16 0
Ac— Nle Asp Val Asn Aib $ Leu Leu $ Leu Lys Val Aib Aib Gln —NH2 17 0
Ac— Nle Asp Val Asn Aib $ Leu Leu $ Leu Lys Val Aib Aib Arg —NH2 18 1
Ac— Met Asp Val Asn Pro Thr $ Leu Phe $ Lys Val Pro Ala Gln —NH2 19 0
Ac— Nle Asp Val Asn Aib Aib $ Leu Phe $ Lys Val Aib Aib Gln —NH2 20 0
Ac— Nle Asp Val Asn Aib Aib $ Leu Phe $ Lys Val Aib Aib Arg —NH2 21 1
Ac— Met Asp Val Asn Pro Thr Leu Leu $ Leu Lys $ Pro Ala Gln —NH2 22 0
Ac— Nle Asp Val Asn Aib Aib Leu Leu $ Leu Lys $ Aib Aib Gln —NH2 23 0
Ac— Nle Asp Val Asn Aib Aib Leu Leu $ Leu Lys $ Aib Aib Arg —NH2 24 1
Ac— Met Asp Val Asn Pro Thr Leu Leu Phe $ Lys Val $ Ala Gln —NH2 25 0
Ac— Nle Asp Val Asn Aib Aib Leu Leu Phe $ Lys Val $ Aib Gln —NH2 26 0
Ac— Nle Asp Val Asn Aib Aib Leu Leu Phe $ Lys Val $ Aib Arg —NH2 27 1
Ac— Met Asp Val Asn Pro Thr Leu Leu Phe Leu Lys $ Pro Ala $ —NH2 28 0
Ac— Nle Asp Val Asn Aib Aib Leu Leu Phe Leu Lys $ Aib Aib $ —NH2 29 0
% Thr Leu Leu Phe Leu Lys Val Pro Ala Gln —NH2 30 1
% Aib Leu Leu Phe Leu Lys Val Aib Aib Gln —NH2 31 1
% Aib Leu Leu Phe Leu Lys Val Aib Aib Arg —NH2 32 2
TABLE 1b
Exemplary PB2 peptidomimetic macrocycles of the invention
Influenza PB2 Sequences For Macrocyclization (bold = critical residue; X = cross-linked amino acid)
1 2 3 4 5 6 7 8 9 10 11 SEQ ID NO: Design Notes
Ac— Met/Nle Glu Arg Ile Lys Glu Leu Arg Asn Leu Met/Nle —NH2 33 wt,
side-chain
H— bonds
contacts
Ac— Met/Nle Glu Arg Ile Lys Glu Leu Arg Asn Leu Met/Nle —NH2 34 wt,
side-chain
hydrophobic
contacts
Ac— X Glu Arg Ile X Glu Leu Arg Asn Leu Met/Nle —NH2 35 i--> i + 4
x-link #1
Ac— Met/Nle Glu Arg Ile X Glu Leu Arg X Leu Met/Nle —NH2 36 i--> i + 4
x-link #2
Ac— Met/Nle X Arg Ile Lys X Leu Arg Asn Leu Met/Nle —NH2 37 i--> i + 4
x-link #3
Ac— X Glu Arg Ile Lys Glu Leu X Asn Leu Met/Nle —NH2 38 i--> i + 7
x-link #1
Ac— Met/Nle X Arg Ile Lys Glu Leu Arg X Leu Met/Nle —NH2 39 i--> i + 7
x-link #2
X Met/Nle Glu XArg He Lys Glu Leu Arg Asn Leu Met/Nle —NH2 40 Formula IV
x-link #1
X Met/Nle Glu XAla Ile Lys Glu Leu Arg Asn Leu Met/Nle —NH2 41 Formula IV
x-link #2
X Met/Nle Glu XGly Ile Lys Glu Leu Arg Asn Leu Met/Nle —NH2 42 Formula IV
x-link #3
Influenza PB2 Peptidomimetic Macrocycles (bold = mutated residue;
S5 = S5-olefin aa; R8 = R8-olefin aa; % = Formula IV x-link) Charge at
1 2 3 4 5 6 7 8 9 10 11 SEQ ID NO: pH 7.4
Ac— S5 Glu Arg Ile S5 Glu Leu Arg Asn Leu Met —NH2 43 0
Ac— Met Glu Arg Ile S5 Glu Leu Arg S5 Leu Met —NH2 44 0
Ac— Met S5 Arg Ile Lys S5 Leu Arg Asn Leu Met —NH2 45 3
Ac— R8 Glu Arg Ile Lys Glu Leu S5 Asn Leu Met —NH2 46 0
Ac— Met R8 Arg Ile Lys Glu Leu Arg S5 Leu Met —NH2 47 2
% Met Glu % Arg Ile Lys Glu Leu Arg Asn Leu Met —NH2 48 1
% Met Glu % Ala Ile Lys Glu Leu Arg Asn Leu Met —NH2 49 0
% Met Glu % Gly Ile Lys Glu Leu Arg Asn Leu Met —NH2 50 0
Ac— S5 Glu Arg Ile S5 Glu Leu Arg Asn Leu Nle —NH2 51 0
Ac— Nle Glu Arg Ile S5 Glu Leu Arg S5 Leu Nle —NH2 52 0
Ac— Nle S5 Arg Ile Lys S5 Leu Arg Asn Leu Nle —NH2 53 3
Ac— R8 Glu Arg Ile Lys Glu Leu S5 Asn Leu Nle —NH2 54 0
Ac— Nle R8 Arg Ile Lys Glu Leu Arg S5 Leu Nle —NH2 55 2
% Nle Glu % Arg Ile Lys Glu Leu Arg Asn Leu Nle —NH2 56 1
% Nle Glu % Ala Ile Lys Glu Leu Arg Asn Leu Nle —NH2 57 0
% Nle Glu % Gly Ile Lys Glu Leu Arg Asn Leu Nle —NH2 58 0
Peptidomimetic Macrocycles of the Invention
In some embodiments, a peptidomimetic macrocycle of the invention has the Formula (I):
Figure US10022422-20180717-C00005
wherein:
each A, C, D, and E is independently a natural or non-natural amino acid;
B is a natural or non-natural amino acid, amino acid analog,
Figure US10022422-20180717-C00006

[—NH-L3-CO—], [—NH-L3-SO2—], or [—NH-L3-];
R1 and R2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-;
R3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5;
L is a macrocycle-forming linker of the formula -L1-L2-;
L1 and L2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R4—K—R4—]n, each being optionally substituted with R5;
each R4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
each K is O, S, SO, SO2, CO, CO2, or CONR3;
each R5 is independently halogen, alkyl, —OR6, —N(R6)2, —SR6, —SOR6, —SO2R6, —CO2R6, a fluorescent moiety, a radioisotope or a therapeutic agent;
each R6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
R7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5, or part of a cyclic structure with a D residue;
R8 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5, or part of a cyclic structure with an E residue;
v and w are independently integers from 1-1000;
u, x, y and z are independently integers from 0-10; and
n is an integer from 1-5.
In one example, at least one of R1 and R2 is alkyl, unsubstituted or substituted with halo-. In another example, both R1 and R2 are independently alkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of R4 and R2 is methyl. In other embodiments, R1 and R2 are methyl.
In some embodiments of the invention, x+y+z is at least 2. In other embodiments of the invention, x+y+z is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. Each occurrence of A, B, C, D or E in a macrocycle or macrocycle precursor of the invention is independently selected. For example, a sequence represented by the formula [A]x, when x is 3, encompasses embodiments where the amino acids are not identical, e.g. Gln-Asp-Ala as well as embodiments where the amino acids are identical, e.g. Gln-Gln-Gln. This applies for any value of x, y, or z in the indicated ranges. Similarly, when u is greater than 1, each compound of the invention may encompass peptidomimetic macrocycles which are the same or different. For example, a compound of the invention may comprise peptidomimetic macrocycles comprising different linker lengths or chemical compositions.
In some embodiments, the peptidomimetic macrocycle of the invention comprises a secondary structure which is a helix and R8 is —H, allowing intrahelical hydrogen bonding. In some embodiments, at least one of A, B, C, D or E is an α,α-disubstituted amino acid. In one example, B is an α,α-disubstituted amino acid. For instance, at least one of A, B, C, D or E is 2-aminoisobutyric acid. In other embodiments, at least one of A, B, C, D or E is
Figure US10022422-20180717-C00007
In other embodiments, the length of the macrocycle-forming linker L as measured from a first Cα to a second Cα is selected to stabilize a desired secondary peptide structure, such as a helix formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first Cα to a second Cα.
In one embodiment, the peptidomimetic macrocycle of Formula (I) is:
Figure US10022422-20180717-C00008
wherein each R1 and R2 is independently H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo.
In related embodiments, the peptidomimetic macrocycle of Formula (I) is:
Figure US10022422-20180717-C00009
Exemplary embodiments of the macrocycle-forming linker L are shown below.
Figure US10022422-20180717-C00010
In some embodiments, the peptidomimetic macrocycles of the invention have the Formula (II):
Figure US10022422-20180717-C00011
wherein:
each A, C, D, and E is independently a natural or non-natural amino acid;
B is a natural or non-natural amino acid, amino acid analog,
Figure US10022422-20180717-C00012

[—NH-L3-CO—], [—NH-L3-SO2—], or [—NH-L3-];
R1 and R2 are independently H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo;
R3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5;
L is a macrocycle-forming linker of the formula
Figure US10022422-20180717-C00013
L1, L2 and L3 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R4—K—R4—]n, each being optionally substituted with R5;
each R4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
each K is O, S, SO, SO2, CO, CO2, or CONR3;
each R5 is independently halogen, alkyl, —OR6, —N(R6)2, —SR6, —SOR6, —SO2R6, —CO2R6, a fluorescent moiety, a radioisotope or a therapeutic agent;
each R6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
R7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5, or part of a cyclic structure with a D residue;
R8 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5, or part of a cyclic structure with an F residue;
v and w are independently integers from 1-1000;
u, x, y and z are independently integers from 0-10; and
n is an integer from 1-5.
In one example, at least one of R1 is alkyl, unsubstituted or substituted with halo-. In another example, both R1 and R2 are independently alkyl, unsubstituted or substituted with halo. In some embodiments, at least one of R1 and R2 is methyl. In other embodiments, R1 and R2 are methyl.
In some embodiments of the invention, x+y+z is at least 2. In other embodiments of the invention, x+y+z is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. Each occurrence of A, B, C, D or E in a macrocycle or macrocycle precursor of the invention is independently selected. For example, a sequence represented by the formula [A]x, when x is 3, encompasses embodiments where the amino acids are not identical, e.g. Gln Asp Ala as well as embodiments where the amino acids are identical, e.g. Gln Gln Gln. This applies for any value of x, y, or z in the indicated ranges.
In some embodiments, the peptidomimetic macrocycle of the invention comprises a secondary structure which is a helix and R8 is —H, allowing intrahelical hydrogen bonding. In some embodiments, at least one of A, B, C, D or F is an α,α-disubstituted amino acid. In one example, B is an α,α-disubstituted amino acid. For instance, at least one of A, B, C, D or E is 2-aminoisobutyric acid. In other embodiments, at least one of A, B, C, D or E is
Figure US10022422-20180717-C00014
In other embodiments, the length of the macrocycle-forming linker L as measured from a first Cα to a second Cα is selected to stabilize a desired secondary peptide structure, such as a helix formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first Cα to a second Cα.
Exemplary embodiments of the macrocycle-forming linker L are shown below.
Figure US10022422-20180717-C00015
Figure US10022422-20180717-C00016
Figure US10022422-20180717-C00017
Figure US10022422-20180717-C00018
Figure US10022422-20180717-C00019
Figure US10022422-20180717-C00020
Figure US10022422-20180717-C00021
Figure US10022422-20180717-C00022
Figure US10022422-20180717-C00023
Figure US10022422-20180717-C00024
In other embodiments, the invention provides peptidomimetic macrocycles of Formula (III):
Figure US10022422-20180717-C00025
wherein:
each A, C, D, and E is independently a natural or non-natural amino acid;
B is a natural or non-natural amino acid, amino acid analog,
Figure US10022422-20180717-C00026

[—NH-L4-CO—], [—NH-L4-SO2—], or [—NH-L4-];
R1 and R2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-;
R3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, unsubstituted or substituted with R5;
L1, L2, L3 and L4 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene or [—R4—K—R4—]n, each being unsubstituted or substituted with R5;
K is O, S, SO, SO2, CO, CO2, or CONR3;
each R4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
each R5 is independently halogen, alkyl, —OR6, —N(R6)2, —SR6, —SOR6, —SO2R6, —CO2R6, a fluorescent moiety, a radioisotope or a therapeutic agent;
each R6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
R7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, unsubstituted or substituted with R5, or part of a cyclic structure with a D residue;
R8 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, unsubstituted or substituted with R5, or part of a cyclic structure with an E residue;
v and w are independently integers from 1-1000;
u, x, y and z are independently integers from 0-10; and
n is an integer from 1-5.
In one example, at least one of R1 and R2 is alkyl, unsubstituted or substituted with halo-. In another example, both R1 and R2 are independently alkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of R1 and R2 is methyl. In other embodiments, R1 and R2 are methyl.
In some embodiments of the invention, x+y+z is at least 2. In other embodiments of the invention, x+y+z is 3, 4, 5, 6, 7, 8, 9 or 10. Each occurrence of A, B, C, D or E in a macrocycle or macrocycle precursor of the invention is independently selected. For example, a sequence represented by the formula [A]x, when x is 3, encompasses embodiments where the amino acids are not identical, e.g. Gln-Asp-Ala as well as embodiments where the amino acids are identical, e.g. Gln-Gln-Gln. This applies for any value of x, y, or z in the indicated ranges.
In some embodiments, the peptidomimetic macrocycle of the invention comprises a secondary structure which is a helix and R8 is —H, allowing intrahelical hydrogen bonding. In some embodiments, at least one of A, B, C, D or E is an α,α-disubstituted amino acid. In one example, B is an α,α-disubstituted amino acid. For instance, at least one of A, B, C, D or E is 2-aminoisobutyric acid. In other embodiments, at least one of A, B, C, D or E is
Figure US10022422-20180717-C00027
In other embodiments, the length of the macrocycle-forming linker [-L1-S-L2-S-L3-] as measured from a first Cα to a second Cα is selected to stabilize a desired secondary peptide structure, such as a helix (including, but not limited to a 310 helix or an α-helix) formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first Cα to a second Cα.
Macrocycles or macrocycle precursors are synthesized, for example, by solution phase or solid-phase methods, and can contain both naturally-occurring and non-naturally-occurring amino acids. See, for example, Hunt, “The Non-Protein Amino Acids” in Chemistry and Biochemistry of the Amino Acids, edited by G. C. Barrett, Chapman and Hall, 1985. In some embodiments, the thiol moieties are the side chains of the amino acid residues L-cysteine, D-cysteine, α-methyl-L cysteine, α-methyl-D-cysteine, L-homocysteine, D-homocysteine, α-methyl-L-homocysteine or α-methyl-D-homocysteine. A bis-alkylating reagent is of the general formula X-L2-Y wherein L2 is a linker moiety and X and Y are leaving groups that are displaced by —SH moieties to form bonds with L2. In some embodiments, X and Y are halogens such as I, Br, or Cl.
In other embodiments, D and/or E in the compound of Formula I, II or III are further modified in order to facilitate cellular uptake. In some embodiments, lipidating or PEGylating a peptidomimetic macrocycle facilitates cellular uptake, increases bioavailability, increases blood circulation, alters pharmacokinetics, decreases immunogenicity and/or decreases the needed frequency of administration.
In other embodiments, at least one of [D] and [E] in the compound of Formula I, II or III represents a moiety comprising an additional macrocycle-forming linker such that the peptidomimetic macrocycle comprises at least two macrocycle-forming linkers. In a specific embodiment, a peptidomimetic macrocycle comprises two macrocycle-forming linkers.
In the peptidomimetic macrocycles of the invention, any of the macrocycle-forming linkers described herein may be used in any combination with any of the sequences shown in Tables 1-4 and also with any of the R substituents indicated herein.
In some embodiments, the peptidomimetic macrocycle of the invention comprises at least one helical motif, such as a 310 or an α-helix motif. For example, A, B and/or C in the compound of Formula I, II or III include one or more helices. As a general matter, helices include between 3 and 4 amino acid residues per turn. In some embodiments, the helix of the peptidomimetic macrocycle includes 1 to 5 turns and, therefore, 3 to 20 amino acid residues. In specific embodiments, the helix includes 1 turn, 2 turns, 3 turns, 4 turns, or 5 turns. In some embodiments, the macrocycle-forming linker stabilizes a helix motif included within the peptidomimetic macrocycle. Thus, in some embodiments, the length of the macrocycle-forming linker L from a first Cα to a second Cα is selected to increase the stability of a helix. In some embodiments, the macrocycle-forming linker spans from 1 turn to 5 turns of the helix. In some embodiments, the macrocycle-forming linker spans approximately 1 turn, 2 turns, 3 turns, 4 turns, or 5 turns of the helix. In some embodiments, the length of the macrocycle-forming linker is approximately 5 Å to 9 Å per turn of the helix, or approximately 6 Å to 8 Å per turn of the helix. Where the macrocycle-forming linker spans approximately 1 turn of a helix, the length is equal to approximately 5 carbon-carbon bonds to 13 carbon-carbon bonds, approximately 7 carbon-carbon bonds to 11 carbon-carbon bonds, or approximately 9 carbon-carbon bonds. Where the macrocycle-forming linker spans approximately 2 turns of a helix, the length is equal to approximately 8 carbon-carbon bonds to 16 carbon-carbon bonds, approximately 10 carbon-carbon bonds to 14 carbon-carbon bonds, or approximately 12 carbon-carbon bonds. Where the macrocycle-forming linker spans approximately 3 turns of a helix, the length is equal to approximately 14 carbon-carbon bonds to 22 carbon-carbon bonds, approximately 16 carbon-carbon bonds to 20 carbon-carbon bonds, or approximately 18 carbon-carbon bonds. Where the macrocycle-forming linker spans approximately 4 turns of a helix, the length is equal to approximately 20 carbon-carbon bonds to 28 carbon-carbon bonds, approximately 22 carbon-carbon bonds to 26 carbon-carbon bonds, or approximately 24 carbon-carbon bonds. Where the macrocycle-forming linker spans approximately 5 turns of a helix, the length is equal to approximately 26 carbon-carbon bonds to 34 carbon-carbon bonds, approximately 28 carbon-carbon bonds to 32 carbon-carbon bonds, or approximately 30 carbon-carbon bonds. Where the macrocycle-forming linker spans approximately 1 turn of a helix, the linkage contains approximately 4 atoms to 12 atoms, approximately 6 atoms to 10 atoms, or approximately 8 atoms. Where the macrocycle-forming linker spans approximately 2 turns of the helix, the linkage contains approximately 7 atoms to 15 atoms, approximately 9 atoms to 13 atoms, or approximately 11 atoms. Where the macrocycle-forming linker spans approximately 3 turns of the helix, the linkage contains approximately 13 atoms to 21 atoms, approximately 15 atoms to 19 atoms, or approximately 17 atoms. Where the macrocycle-forming linker spans approximately 4 turns of the helix, the linkage contains approximately 19 atoms to 27 atoms, approximately 21 atoms to 25 atoms, or approximately 23 atoms. Where the macrocycle-forming linker spans approximately 5 turns of the helix, the linkage contains approximately 25 atoms to 33 atoms, approximately 27 atoms to 31 atoms, or approximately 29 atoms. Where the macrocycle-forming linker spans approximately 1 turn of the helix, the resulting macrocycle forms a ring containing approximately 17 members to 25 members, approximately 19 members to 23 members, or approximately 21 members. Where the macrocycle-forming linker spans approximately 2 turns of the helix, the resulting macrocycle forms a ring containing approximately 29 members to 37 members, approximately 31 members to 35 members, or approximately 33 members. Where the macrocycle-forming linker spans approximately 3 turns of the helix, the resulting macrocycle forms a ring containing approximately 44 members to 52 members, approximately 46 members to 50 members, or approximately 48 members. Where the macrocycle-forming linker spans approximately 4 turns of the helix, the resulting macrocycle forms a ring containing approximately 59 members to 67 members, approximately 61 members to 65 members, or approximately 63 members. Where the macrocycle-forming linker spans approximately 5 turns of the helix, the resulting macrocycle forms a ring containing approximately 74 members to 82 members, approximately 76 members to 80 members, or approximately 78 members.
In other embodiments, the invention provides peptidomimetic macrocycles of Formula (IV) or (IVa):
Figure US10022422-20180717-C00028
wherein:
each A, C, D, and E is independently a natural or non-natural amino acid;
B is a natural or non-natural amino acid, amino acid analog,
Figure US10022422-20180717-C00029

[—NH-L3-CO—], [—NH-L3-SO2—], or [—NH-L3-];
R1 and R2 are independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-, or part of a cyclic structure with an E residue;
R3 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5;
L is a macrocycle-forming linker of the formula -L4-L2-;
L1 and L2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R4—K—R4—]n, each being optionally substituted with R5;
each R4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
each K is O, S, SO, SO2, CO, CO2, or CONR3;
each R5 is independently halogen, alkyl, —OR6, —N(R6)2, —SR6, —SOR6, —SO2R6, —CO2R6, a fluorescent moiety, a radioisotope or a therapeutic agent;
each R6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent;
R7 is —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, heteroalkyl, cycloalkylalkyl, heterocycloalkyl, cycloaryl, or heterocycloaryl, optionally substituted with R5;
v and w are independently integers from 1-1000;
u, x, y and z are independently integers from 0-10; and
n is an integer from 1-5.
In one example, at least one of R1 and R2 is alkyl, unsubstituted or substituted with halo-. In another example, both R1 and R2 are independently alkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of R1 and R2 is methyl. In other embodiments, R1 and R2 are methyl.
In some embodiments of the invention, x+y+z is at least 1. In other embodiments of the invention, x+y+z is at least 2. In other embodiments of the invention, x+y+z is 3, 4, 5, 6, 7, 8, 9 or 10. Each occurrence of A, B, C, D or E in a macrocycle or macrocycle precursor of the invention is independently selected. For example, a sequence represented by the formula [A]x, when x is 3, encompasses embodiments where the amino acids are not identical, e.g. Gln-Asp-Ala as well as embodiments where the amino acids are identical, e.g. Gln-Gln-Gln. This applies for any value of x, y, or z in the indicated ranges.
In some embodiments, the peptidomimetic macrocycle of the invention comprises a secondary structure which is a helix and R is —H, allowing intrahelical hydrogen bonding. In some embodiments, at least one of A, B, C, D or E is an α,α-disubstituted amino acid. In one example, 13 is an α,α-disubstituted amino acid. For instance, at least one of A, B, C, D or E is 2-aminoisobutyric acid. In other embodiments, at least one of A, B, C, D or E is
Figure US10022422-20180717-C00030
In other embodiments, the length of the macrocycle-forming linker L as measured from a first Cα to a second Cα is selected to stabilize a desired secondary peptide structure, such as a helix (including a 310 helix or α-helix) formed by residues of the peptidomimetic macrocycle including, but not necessarily limited to, those between the first Cα to a second Cα.
Exemplary embodiments of the macrocycle-forming linker -L1-L2- are shown below.
Figure US10022422-20180717-C00031

Preparation of Peptidomimetic Macrocycles
Peptidomimetic macrocycles of the invention may be prepared by any of a variety of methods known in the art. For example, any of the residues indicated by “X” in Table 1 may be substituted with a residue capable of forming a crosslinker with a second residue in the same molecule or a precursor of such a residue.
Various methods to effect formation of peptidomimetic macrocycles are known in the art. For example, the preparation of peptidomimetic macrocycles of Formula I is described in Schafmeister et al., J. Am. Chem. Soc. 122:5891-5892 (2000); Schafmeister & Verdine, J. Am. Chem. Soc. 122:5891 (2005); Walensky et al, Science 305:1466-1470 (2004); and U.S. Pat. No. 7,192,713. The α,α-disubstituted amino acids and amino acid precursors disclosed in the cited references may be employed in synthesis of the peptidomimetic macrocycle precursor polypeptides. For example, the “S5-olefin amino acid” is (S)-α-(2′-pentenyl) alanine and the “R8 olefin amino acid” is (R)-α-(2′-octenyl) alanine. Following incorporation of such amino acids into precursor polypeptides, the terminal olefins are reacted with a metathesis catalyst, leading to the formation of the peptidomimetic macrocycle.
In other embodiments, the peptidomimetic macrocycles of the invention are of Formula IV or IVa. Methods for the preparation of such macrocycles are described, for example, in U.S. Pat. No. 7,202,332.
In some embodiments, the synthesis of these peptidomimetic macrocycles involves a multi-step process that features the synthesis of a peptidomimetic precursor containing an azide moiety and an alkyne moiety; followed by contacting the peptidomimetic precursor with a macrocyclization reagent to generate a triazole-linked peptidomimetic macrocycle. Such a process is described, for example, in U.S. application Ser. No. 12/037,041, filed on Feb. 25, 2008. Macrocycles or macrocycle precursors are synthesized, for example, by solution phase or solid-phase methods, and can contain both naturally-occurring and non-naturally-occurring amino acids. See, for example, Hunt, “The Non-Protein Amino Acids” in Chemistry and Biochemistry of the Amino Acids, edited by G. C. Barrett, Chapman and Hall, 1985.
In some embodiments, an azide is linked to the α-carbon of a residue and an alkyne is attached to the α-carbon of another residue. In some embodiments, the azide moieties are azido-analogs of amino acids L-lysine, D-lysine, alpha-methyl-L-lysine, alpha-methyl-D-lysine, L-omithine, D-ornithine, alpha-methyl-L-omithine or alpha-methyl-D-ornithine. In another embodiment, the alkyne moiety is L-propargylglycine. In yet other embodiments, the alkyne moiety is an amino acid selected from the group consisting of L-propargylglycine, D-propargylglycine, (S)-2-amino-2-methyl-4-pentynoic acid, (R)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-2-methyl-5-hexynoic acid, (R)-2-amino-2-methyl-5-hexynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, (R)-2-amino-2-methyl-6-heptynoic acid, (S)-2-amino-2-methyl-7-octynoic acid, (R)-2-amino-2-methyl-7-octynoic acid, (S)-2-amino-2-methyl-8-nonynoic acid and (R)-2-amino-2-methyl-8-nonynoic acid.
In some embodiments, the invention provides a method for synthesizing a peptidomimetic macrocycle, the method comprising the steps of contacting a peptidomimetic precursor of Formula V or Formula VI:
Figure US10022422-20180717-C00032
with a macrocyclization reagent;
wherein v, w, x, y, z, A, B, C, D, E, R1, R2, R7, R8, L1 and L2 are as defined for Formula (II); R12 is —H when the macrocyclization reagent is a Cu reagent and R12 is —H or alkyl when the macrocyclization reagent is a Ru reagent; and further wherein said contacting step results in a covalent linkage being formed between the alkyne and azide moiety in Formula III or Formula IV. For example, R12 may be methyl when the macrocyclization reagent is a Ru reagent.
In the peptidomimetic macrocycles of the invention, at least one of R1 and R2 is alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-. In some embodiments, both R1 and R2 are independently alkyl, alkenyl, alkynyl, arylalkyl, cycloalkyl, cycloalkylalkyl, heteroalkyl, or heterocycloalkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of A, B, C, D or E is an α,α-disubstituted amino acid. In one example, B is an α,α-disubstituted amino acid. For instance, at least one of A, B, C, D or E is 2-aminoisobutyric acid.
For example, at least one of R1 and R2 is alkyl, unsubstituted or substituted with halo-. In another example, both R1 and R2 are independently alkyl, unsubstituted or substituted with halo-. In some embodiments, at least one of R1 and R2 is methyl. In other embodiments, R1 and R2 are methyl. The macrocyclization reagent may be a Cu reagent or a Ru reagent.
In some embodiments, the peptidomimetic precursor is purified prior to the contacting step. In other embodiments, the peptidomimetic macrocycle is purified after the contacting step. In still other embodiments, the peptidomimetic macrocycle is refolded after the contacting step. The method may be performed in solution, or, alternatively, the method may be performed on a solid support.
Also envisioned herein is performing the method of the invention in the presence of a target macromolecule that binds to the peptidomimetic precursor or peptidomimetic macrocycle under conditions that favor said binding. In some embodiments, the method is performed in the presence of a target macromolecule that binds preferentially to the peptidomimetic precursor or peptidomimetic macrocycle under conditions that favor said binding. The method may also be applied to synthesize a library of peptidomimetic macrocycles.
In some embodiments, the alkyne moiety of the peptidomimetic precursor of Formula V or Formula VI is a sidechain of an amino acid selected from the group consisting of L-propargylglycine, D-propargylglycine, (S)-2-amino-2-methyl-4-pentynoic acid, (R)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-2-methyl-5-hexynoic acid, (R)-2-amino-2-methyl-5-hexynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, (R)-2-amino-2-methyl-6-heptynoic acid, (S)-2-amino-2-methyl-7-octynoic acid, (R)-2-amino-2-methyl-7-octynoic acid, (S)-2-amino-2-methyl-8-nonynoic acid, and (R)-2-amino-2-methyl-8-nonynoic acid. In other embodiments, the azide moiety of the peptidomimetic precursor of Formula V or Formula VI is a sidechain of an amino acid selected from the group consisting of ε-azido-L-lysine, ε-azido-D-lysine, ε-azido-α-methyl-L-lysine, ε-azido-α-methyl-D-lysine, δ-azido-α-methyl-L-omithine, and δ-azido-α-methyl-D-ornithine.
In some embodiments, x+y+z is 2, and A, B and C are independently natural or non-natural amino acids. In other embodiments, x+y+z is 3 or 6, and A, B and C are independently natural or non-natural amino acids.
In some embodiments, the contacting step is performed in a solvent selected from the group consisting of protic solvent, aqueous solvent, organic solvent, and mixtures thereof. For example, the solvent may be chosen from the group consisting of H2O, THF, THF/H2O, tBuOH/H2O, DMF, DIPEA, CH3CN or CH2Cl2, ClCH2CH2Cl or a mixture thereof. The solvent may be a solvent which favors helix formation.
Alternative but equivalent protecting groups, leaving groups or reagents are substituted, and certain of the synthetic steps are performed in alternative sequences or orders to produce the desired compounds. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein include, for example, those such as described in Larock, Comprehensive Organic Transformations, VCH Publishers (1989); Greene and Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); Fieser and Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof.
In some embodiments, peptidomimetic macrocycles of the invention have the structure (SEQ ID NOS 59-62, respectively, in order of appearance):
Figure US10022422-20180717-C00033
The influenza PB1 peptidomimetic macrocycles shown above are also identified as SP-8, SP-16, SP-13 and SP-41, respectively. In other embodiments, the peptidomimetic macrocycle of the invention has the structure (SEQ ID NOS 63-65, respectively, in order of appearance):
Figure US10022422-20180717-C00034
Preparation of Peptidomimetic Macrocycles.
The peptidomimetic macrocycles of the invention are made, for example, by chemical synthesis methods, such as described in Fields et al., Chapter 3 in Synthetic Peptides: A User's Guide, ed. Grant, W. H. Freeman & Co., New York, N.Y., 1992, p. 77. Hence, for example, peptides are synthesized using the automated Merrifield techniques of solid phase synthesis with the amine protected by either tBoc or Fmoc chemistry using side chain protected amino acids on, for example, an automated peptide synthesizer (e.g., Applied Biosystems (Foster City, Calif.), Model 430A, 431, or 433).
One manner of producing the peptidomimetic precursors and peptidomimetic macrocycles described herein uses solid phase peptide synthesis (SPPS). The C-terminal amino acid is attached to a cross-linked polystyrene resin via an acid labile bond with a linker molecule. This resin is insoluble in the solvents used for synthesis, making it relatively simple and fast to wash away excess reagents and by-products. The N-terminus is protected with the Fmoc group, which is stable in acid, but removable by base. Side chain functional groups are protected as necessary with base stable, acid labile groups.
Longer peptidomimetic precursors are produced, for example, by conjoining individual synthetic peptides using native chemical ligation. Alternatively, the longer synthetic peptides are biosynthesized by well known recombinant DNA and protein expression techniques. Such techniques are provided in well-known standard manuals with detailed protocols. To construct a gene encoding a peptidomimetic precursor of this invention, the amino acid sequence is reverse translated to obtain a nucleic acid sequence encoding the amino acid sequence, preferably with codons that are optimum for the organism in which the gene is to be expressed. Next, a synthetic gene is made, typically by synthesizing oligonucleotides which encode the peptide and any regulatory elements, if necessary. The synthetic gene is inserted in a suitable cloning vector and transfected into a host cell. The peptide is then expressed under suitable conditions appropriate for the selected expression system and host. The peptide is purified and characterized by standard methods.
The peptidomimetic precursors are made, for example, in a high-throughput, combinatorial fashion using, for example, a high-throughput polychannel combinatorial synthesizer (e.g., Thuramed TETRAS multichannel peptide synthesizer from CreoSalus, Louisville, Ky. or Model Apex 396 multichannel peptide synthesizer from AAPPTEC, Inc., Louisville, Ky.).
The following synthetic schemes are provided solely to illustrate the present invention and are not intended to limit the scope of the invention, as described herein. To simplify the drawings, the illustrative schemes depict azido amino acid analogs ε-azido-α-methyl-L-lysine and ε-azido-α-methyl-D-lysine, and alkyne amino acid analogs L-propargylglycine, (S)-2-amino-2-methyl-4-pentynoic acid, and (S)-2-amino-2-methyl-6-heptynoic acid. Thus, in the following synthetic schemes, each R1, R2, R7 and R8 is —H; each L1 is —(CH2)4—; and each L2 is —(CH2)—. However, as noted throughout the detailed description above, many other amino acid analogs can be employed in which R1, R2, R7, R8, L1 and L2 can be independently selected from the various structures disclosed herein.
Figure US10022422-20180717-C00035
Figure US10022422-20180717-C00036
Synthetic Scheme 1 describes the preparation of several compounds of the invention. Ni(II) complexes of Schiff bases derived from the chiral auxiliary (S)-2-[N—(N′-benzylpropyl)amino]benzophenone (BPB) and amino acids such as glycine or alanine are prepared as described in Belokon et al. (1998), Tetrahedron Asymm. 9:4249-4252. The resulting complexes are subsequently reacted with alkylating reagents comprising an azido or alkynyl moiety to yield enantiomerically enriched compounds of the invention. If desired, the resulting compounds can be protected for use in peptide synthesis.
Figure US10022422-20180717-C00037
In the general method for the synthesis of peptidomimetic macrocycles shown in Synthetic Scheme 2, the peptidomimetic precursor contains an azide moiety and an alkyne moiety and is synthesized by solution-phase or solid-phase peptide synthesis (SPPS) using the commercially available amino acid N-α-Fmoc-L-propargylglycine and the N-α-Fmoc-protected forms of the amino acids (S)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-6-heptynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, N-methyl-ε-azido-L-lysine, and N-methyl-ε-azido-D-lysine. The peptidomimetic precursor is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA). The peptidomimetic precursor is reacted as a crude mixture or is purified prior to reaction with a macrocyclization reagent such as a Cu(I) in organic or aqueous solutions (Rostovtsev et al. (2002), Angew. Chem. Int. Ed. 41:2596-2599; Tornoe et al. (2002), J. Org. Chem. 67:3057-3064; Deiters et al. (2003), J. Am. Chem. Soc. 125:11782-11783; Punna et al. (2005), Angew. Chem. Int. Ed. 44:2215-2220). In one embodiment, the triazole forming reaction is performed under conditions that favor helix formation. In one embodiment, the macrocyclization step is performed in a solvent chosen from the group consisting of H2O, THF, CH3CN, DMF, DIPEA, tBuOH or a mixture thereof. In another embodiment, the macrocyclization step is performed in DMF. In some embodiments, the macrocyclization step is performed in a buffered aqueous or partially aqueous solvent.
Figure US10022422-20180717-C00038
In the general method for the synthesis of peptidomimetic macrocycles shown in Synthetic Scheme 3, the peptidomimetic precursor contains an azide moiety and an alkyne moiety and is synthesized by solid-phase peptide synthesis (SPPS) using the commercially available amino acid N-α-Fmoc-L-propargylglycine and the N-α-Fmoc-protected forms of the amino acids (S)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-6-heptynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, N-methyl-ε-azido-L-lysine, and N-methyl-ε-azido-D-lysine. The peptidomimetic precursor is reacted with a macrocyclization reagent such as a Cu(I) reagent on the resin as a crude mixture (Rostovtsev et al. (2002), Angew. Chem. Int. Ed. 41:2596-2599; Tornoe et al. (2002), J. Org. Chem. 67:3057-3064; Deiters et al. (2003), J. Am. Chem. Soc. 125:11782-11783; Punna et al. (2005), Angew. Chem. Int. Ed. 44:2215-2220). The resultant triazole-containing peptidomimetic macrocycle is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA). In some embodiments, the macrocyclization step is performed in a solvent chosen from the group consisting of CH2Cl2, ClCH2CH2Cl, DMF, THF, NMP, DIPEA, 2,6-lutidine, pyridine, DMSO, H2O or a mixture thereof. In some embodiments, the macrocyclization step is performed in a buffered aqueous or partially aqueous solvent.
Figure US10022422-20180717-C00039
In the general method for the synthesis of peptidomimetic macrocycles shown in Synthetic Scheme 4, the peptidomimetic precursor contains an azide moiety and an alkyne moiety and is synthesized by solution-phase or solid-phase peptide synthesis (SPPS) using the commercially available amino acid N-α-Fmoc-L-propargylglycine and the N-α-Fmoc-protected forms of the amino acids (S)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-6-heptynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, N-methyl-ε-azido-L-lysine, and N-methyl-ε-azido-D-lysine. The peptidomimetic precursor is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA). The peptidomimetic precursor is reacted as a crude mixture or is purified prior to reaction with a macrocyclization reagent such as a Ru(II) reagents, for example Cp*RuCl(PPh3)2 or [Cp*RuCl]4 (Rasmussen et al. (2007), Org. Lett. 9:5337-5339; Zhang et al. (2005), J. Am. Chem. Soc. 127:15998-15999). In some embodiments, the macrocyclization step is performed in a solvent chosen from the group consisting of DMF, CH3CN and THF.
Figure US10022422-20180717-C00040
In the general method for the synthesis of peptidomimetic macrocycles shown in Synthetic Scheme 5, the peptidomimetic precursor contains an azide moiety and an alkyne moiety and is synthesized by solid-phase peptide synthesis (SPPS) using the commercially available amino acid N-α-Fmoc-L-propargylglycine and the N-α-Fmoc-protected forms of the amino acids (S)-2-amino-2-methyl-4-pentynoic acid, (S)-2-amino-6-heptynoic acid, (S)-2-amino-2-methyl-6-heptynoic acid, N-methyl-ε-azido-L-lysine, and N-methyl-ε-azido-D-lysine. The peptidomimetic precursor is reacted with a macrocyclization reagent such as a Ru(H) reagent on the resin as a crude mixture. For example, the reagent can be Cp*RuCl(PPh3)2 or [Cp*RuCl]4 (Rasmussen et al. (2007), Org. Lett. 9:5337-5339; Zhang et al. (2005), J. Am. Chem. Soc. 127:15998-15999). In some embodiments, the macrocyclization step is performed in a solvent chosen from the group consisting of CH2Cl2, ClCH2CH2Cl, CH3CN, DMF, and THF.
The present invention contemplates the use of non-naturally-occurring amino acids and amino acid analogs in the synthesis of the peptidomimetic macrocycles described herein. Any amino acid or amino acid analog amenable to the synthetic methods employed for the synthesis of stable triazole containing peptidomimetic macrocycles can be used in the present invention. For example, L-propargylglycine is contemplated as a useful amino acid in the present invention. However, other alkyne-containing amino acids that contain a different amino acid side chain are also useful in the invention. For example, L-propargylglycine contains one methylene unit between the α-carbon of the amino acid and the alkyne of the amino acid side chain. The invention also contemplates the use of amino acids with multiple methylene units between the α-carbon and the alkyne. Also, the azido-analogs of amino acids L-lysine, D-lysine, alpha-methyl-L-lysine, and alpha-methyl-D-lysine are contemplated as useful amino acids in the present invention. However, other terminal azide amino acids that contain a different amino acid side chain are also useful in the invention. For example, the azido-analog of L-lysine contains four methylene units between the α-carbon of the amino acid and the terminal azide of the amino acid side chain. The invention also contemplates the use of amino acids with fewer than or greater than four methylene units between the α-carbon and the terminal azide. Table 2 shows some amino acids useful in the preparation of peptidomimetic macrocycles of the invention.
TABLE 2
Figure US10022422-20180717-C00041
N-α-Fmoc-L-propargyl glycine
Figure US10022422-20180717-C00042
N-α-Fmoc-D-propargyl glycine
Figure US10022422-20180717-C00043
N-α-Fmoc-(S)-2-amino-2-methyl-4-pentynoic acid
Figure US10022422-20180717-C00044
N-α-Fmoc-(R)-2-amino-2-methyl-4-pentynoic acid
Figure US10022422-20180717-C00045
N-α-Fmoc-ϵ-azido-L-lysine
Figure US10022422-20180717-C00046
N-α-Fmoc-ϵ-azido-D-lysine
Figure US10022422-20180717-C00047
N-α-Fmoc-(S)-2-amino-2-methyl-5-hexynoic acid
Figure US10022422-20180717-C00048
N-α-Fmoc-(R)-2-amino-2-methyl-5-hexynoic acid
Figure US10022422-20180717-C00049
N-α-Fmoc-ϵ-azido-α-methyl-L-lysine
Figure US10022422-20180717-C00050
N-α-Fmoc-ϵ-azido-α-methyl-D-lysine
Figure US10022422-20180717-C00051
N-α-Fmoc-(S)-2-amino-2-methyl-6-heptynoic acid
Figure US10022422-20180717-C00052
N-α-Fmoc-(R)-2-amino-2-methyl-6-heptynoic acid
Figure US10022422-20180717-C00053
N-α-Fmoc-(S)-2-amino-2-methyl-7-octynoic acid
Figure US10022422-20180717-C00054
N-α-Fmoc-(R)-2-amino-2-methyl-7-octynoic acid
Figure US10022422-20180717-C00055
N-α-Fmoc-δ-azido-L-ornithine
Figure US10022422-20180717-C00056
N-α-Fmoc-δ-azido-D-ornithine
Figure US10022422-20180717-C00057
N-α-Fmoc-(S)-2-amino-2-methyl-8-nonynoic acid
Figure US10022422-20180717-C00058
N-α-Fmoc-(R)-2-amino-2-methyl-8-nonynoic acid
Figure US10022422-20180717-C00059
N-α-Fmoc-ϵ-azido-α-methyl-L-ornithine
Figure US10022422-20180717-C00060
N-α-Fmoc-ϵ-azido-α-methyl-D-ornithine
Table 2 shows exemplary amino acids useful in the preparation of peptidomimetic macrocycles of the invention.
In some embodiments the amino acids and amino acid analogs are of the D-configuration. In other embodiments they are of the L-configuration. In some embodiments, some of the amino acids and amino acid analogs contained in the peptidomimetic are of the D-configuration while some of the amino acids and amino acid analogs are of the L-configuration. In some embodiments the amino acid analogs are α,α-disubstituted, such as α-methyl-L-propargylglycine, α-methyl-D-propargylglycine, ε-azido-alpha-methyl-L-lysine, and ε-azido-alpha-methyl-D-lysine. In some embodiments the amino acid analogs are N-alkylated, e.g., N-methyl-L-propargylglycine, N-methyl-D-propargylglycine, N-methyl-ε-azido-L-lysine, and N-methyl-ε-azido-D-lysine.
In some embodiments, the —NH moiety of the amino acid is protected using a protecting group, including without limitation -Fmoc and -Boc. In other embodiments, the amino acid is not protected prior to synthesis of the peptidomimetic macrocycle.
In other embodiments, peptidomimetic macrocycles of Formula III are synthesized. The preparation of such macrocycles is described, for example, in U.S. application Ser. No. 11/957,325, filed on Dec. 17, 2007. The following synthetic schemes describe the preparation of such compounds. To simplify the drawings, the illustrative schemes depict amino acid analogs derived from L- or D-cysteine, in which L1 and L3 are both —(CH2)—. However, as noted throughout the detailed description above, many other amino acid analogs can be employed in which L1 and L3 can be independently selected from the various structures disclosed herein. The symbols “[AA]m”, “[AA]n”, “[AA]o” represent a sequence of amide bond-linked moieties such as natural or unnatural amino acids. As described previously, each occurrence of “AA” is independent of any other occurrence of “AA”, and a formula such as “[AA]m” encompasses, for example, sequences of non-identical amino acids as well as sequences of identical amino acids.
Figure US10022422-20180717-C00061
In Scheme 6, the peptidomimetic precursor contains two —SH moieties and is synthesized by solid-phase peptide synthesis (SPPS) using commercially available N-α-Fmoc amino acids such as N-α-Fmoc-S-trityl-L-cysteine or N-α-Fmoc-S-trityl-D-cysteine. Alpha-methylated versions of D-cysteine or L-cysteine are generated by known methods (Seebach et al. (1996), Angew. Chem. Int. Ed. Engl. 35:2708-2748, and references therein) and then converted to the appropriately protected N-α-Fmoc-S-trityl monomers by known methods (“Bioorganic Chemistry: Peptides and Proteins”, Oxford University Press, New York: 1998, the entire contents of which are incorporated herein by reference). The precursor peptidomimetic is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA). The precursor peptidomimetic is reacted as a crude mixture or is purified prior to reaction with X-L2-Y in organic or aqueous solutions. In some embodiments the alkylation reaction is performed under dilute conditions (i.e. 0.15 mmol/L) to favor macrocyclization and to avoid polymerization. In some embodiments, the alkylation reaction is performed in organic solutions such as liquid NH3 (Mosberg et al. (1985), J. Am. Chem. Soc. 107:2986-2987; Szewczuk et al. (1992), Int. J. Peptide Protein Res. 40: 233-242), NH3/MeOH, or NH3/DMF (Or et al. (1991), J. Org. Chem. 56:3146-3149). In other embodiments, the alkylation is performed in an aqueous solution such as 6M guanidinium HCL, pH 8 (Brunel et al. (2005), Chem. Commun. (20):2552-2554). In other embodiments, the solvent used for the alkylation reaction is DMF or
Figure US10022422-20180717-C00062
In Scheme 7, the precursor peptidomimetic contains two or more —SH moieties, of which two are specially protected to allow their selective deprotection and subsequent alkylation for macrocycle formation. The precursor peptidomimetic is synthesized by solid-phase peptide synthesis (SPPS) using commercially available N-α-Fmoc amino acids such as N-α-Fmoc-S-p-methoxytrityl-L-cysteine or N-α-Fmoc-S-p-methoxytrityl-D-cysteine. Alpha-methylated versions of D-cysteine or L-cysteine are generated by known methods (Seebach et al. (1996), Angew. Chem. Int. Ed. Engl. 35:2708-2748, and references therein) and then converted to the appropriately protected N-α-Fmoc-S-p-methoxytrityl monomers by known methods (Bioorganic Chemistry: Peptides and Proteins, Oxford University Press, New York: 1998, the entire contents of which are incorporated herein by reference). The Mmt protecting groups of the peptidomimetic precursor are then selectively cleaved by standard conditions (e.g., mild acid such as 1% TFA in DCM). The precursor peptidomimetic is then reacted on the resin with X-L2-Y in an organic solution. For example, the reaction takes place in the presence of a hindered base such as diisopropylethylamine. In some embodiments, the alkylation reaction is performed in organic solutions such as liquid NH3 (Mosberg et al. (1985), J. Am. Chem. Soc. 107:2986-2987; Szewczuk et al. (1992), Int. J. Peptide Protein Res. 40:233-242), NH3/MeOH or NH3/DMF (Or et al. (1991), J. Org. Chem. 56:3146-3149). In other embodiments, the alkylation reaction is performed in DMF or dichloroethane. The peptidomimetic macrocycle is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA).
Figure US10022422-20180717-C00063
In Scheme 8, the peptidomimetic precursor contains two or more —SH moieties, of which two are specially protected to allow their selective deprotection and subsequent alkylation for macrocycle formation. The peptidomimetic precursor is synthesized by solid-phase peptide synthesis (SPPS) using commercially available N-α-Fmoc amino acids such as N-α-Fmoc-S-p-methoxytrityl-L-cysteine, N-α-Fmoc-S-p-methoxytrityl-D-cysteine, N-α-Fmoc-S—S-t-butyl-L-cysteine, and N-α-Fmoc-S—S-t-butyl-D-cysteine. Alpha-methylated versions of D-cysteine or L-cysteine are generated by known methods (Secbach et al. (1996), Angew. Chem. Int. Ed. Engl. 35:2708-2748, and references therein) and then converted to the appropriately protected N-α-Fmoc-S-p-methoxytrityl or N-α-Fmoc-S—S-t-butyl monomers by known methods (Bioorganic Chemistry: Peptides and Proteins, Oxford University Press, New York: 1998, the entire contents of which are incorporated herein by reference). The S—S-tButyl protecting group of the peptidomimetic precursor is selectively cleaved by known conditions (e.g., 20% 2-mercaptoethanol in DMF, reference: Galande et al. (2005), J. Comb. Chem. 7:174-177). The precursor peptidomimetic is then reacted on the resin with a molar excess of X-L2-Y in an organic solution. For example, the reaction takes place in the presence of a hindered base such as diisopropylethylamine. The Mmt protecting group of the peptidomimetic precursor is then selectively cleaved by standard conditions (e.g., mild acid such as 1% TFA in DCM). The peptidomimetic precursor is then cyclized on the resin by treatment with a hindered base in organic solutions. In some embodiments, the alkylation reaction is performed in organic solutions such as NH3/MeOH or NH3/DMF (Or et al. (1991), J. Org. Chem. 56:3146-3149). The peptidomimetic macrocycle is then deprotected and cleaved from the solid-phase resin by standard conditions (e.g., strong acid such as 95% TFA).
Figure US10022422-20180717-C00064
In Scheme 9, the peptidomimetic precursor contains two L-cysteine moieties. The peptidomimetic precursor is synthesized by known biological expression systems in living cells or by known in vitro, cell-free, expression methods. The precursor peptidomimetic is reacted as a crude mixture or is purified prior to reaction with X-L2-Y in organic or aqueous solutions. In some embodiments the alkylation reaction is performed under dilute conditions (i.e. 0.15 mmol/L) to favor macrocyclization and to avoid polymerization. In some embodiments, the alkylation reaction is performed in organic solutions such as liquid NH3 (Mosberg et al. (1985), J. Am. Chem. Soc. 107:2986-2987; Szewczuk et al. (1992), Int. J. Peptide Protein Res. 40:233-242), NH3/MeOH, or NH3/DMF (Or et al. (1991), J. Org. Chem. 56:3146-3149). In other embodiments, the alkylation is performed in an aqueous solution such as 6M guanidinium HCL, pH 8 (Brunel et al. (2005), Chem. Commun. (20):2552-2554). In other embodiments, the alkylation is performed in DMF or dichloroethane. In another embodiment, the alkylation is performed in non-denaturing aqueous solutions, and in yet another embodiment the alkylation is performed under conditions that favor helical structure formation. In yet another embodiment, the alkylation is performed under conditions that favor the binding of the precursor peptidomimetic to another protein, so as to induce the formation of the bound helical conformation during the alkylation.
Various embodiments for X and Y are envisioned which are suitable for reacting with thiol groups. In general, each X or Y is independently be selected from the general category shown in Table 5. For example, X and Y are halides such as —Cl, —Br or —I. Any of the macrocycle-forming linkers described herein may be used in any combination with any of the sequences shown in Tables 1-4 and also with any of the R-substituents indicated herein.
TABLE 3
Examples of Reactive Groups Capable of Reacting
with Thiol Groups and Resulting Linkages
Resulting
X or Y Covalent Linkage
acrylamide Thioether
halide (e.g. alkyl or aryl halide) Thioether
sulfonate Thioether
aziridine Thioether
epoxide Thioether
haloacetamide Thioether
maleimide Thioether
sulfonate ester Thioether
The present invention contemplates the use of both naturally-occurring and non-naturally-occurring amino acids and amino acid analogs in the synthesis of the peptidomimetic macrocycles of Formula (III). Any amino acid or amino acid analog amenable to the synthetic methods employed for the synthesis of stable bis-sulfhydryl containing peptidomimetic macrocycles can be used in the present invention. For example, cysteine is contemplated as a useful amino acid in the present invention. However, sulfur containing amino acids other than cysteine that contain a different amino acid side chain are also useful. For example, cysteine contains one methylene unit between the α-carbon of the amino acid and the terminal —SH of the amino acid side chain. The invention also contemplates the use of amino acids with multiple methylene units between the α-carbon and the terminal —SH. Non-limiting examples include α-methyl-L-homocysteine and α-methyl-D-homocysteine. In some embodiments the amino acids and amino acid analogs are of the D-configuration. In other embodiments they are of the L-configuration. In some embodiments, some of the amino acids and amino acid analogs contained in the peptidomimetic are of the D-configuration while some of the amino acids and amino acid analogs are of the L-configuration. In some embodiments the amino acid analogs are α,α-disubstituted, such as α-methyl-L-cysteine and α-methyl-D-cysteine.
The invention includes macrocycles in which macrocycle-forming linkers are used to link two or more —SH moieties in the peptidomimetic precursors to form the peptidomimetic macrocycles of the invention. As described above, the macrocycle-forming linkers impart conformational rigidity, increased metabolic stability and/or increased cell penetrability. Furthermore, in some embodiments, the macrocycle-forming linkages stabilize a helical secondary structure of the peptidomimetic macrocyles. The macrocycle-forming linkers are of the formula X-L2-Y, wherein both X and Y are the same or different moieties, as defined above. Both X and Y have the chemical characteristics that allow one macrocycle-forming linker -L2- to bis alkylate the bis-sulfhydryl containing peptidomimetic precursor. As defined above, the linker -L2- includes alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, or heterocycloarylene, or —R4—K—R4—, all of which can be optionally substituted with an R5 group, as defined above. Furthermore, one to three carbon atoms within the macrocycle-forming linkers other than the carbons attached to the —SH of the sulfhydryl containing amino acid, are optionally substituted with a heteroatom such as N, S or O.
The L2 component of the macrocycle-forming linker X-L2-Y may be varied in length depending on, among other things, the distance between the positions of the two amino acid analogs used to form the peptidomimetic macrocycle. Furthermore, as the lengths of L1 and/or L3 components of the macrocycle-forming linker are varied, the length of L2 can also be varied in order to create a linker of appropriate overall length for forming a stable peptidomimetic macrocycle. For example, if the amino acid analogs used are varied by adding an additional methylene unit to each of L1 and L3, the length of L2 are decreased in length by the equivalent of approximately two methylene units to compensate for the increased lengths of L1 and L3.
In some embodiments, L2 is an alkylene group of the formula —(CH2)n—, where n is an integer between about 1 and about 15. For example, n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. In other embodiments, L2 is an alkenylene group. In still other embodiments, L2 is an aryl group.
Table 4 shows additional embodiments of X-L2-Y groups.
TABLE 4
Exemplary X—L2—Y groups of the invention.
Figure US10022422-20180717-C00065
Figure US10022422-20180717-C00066
Figure US10022422-20180717-C00067
Figure US10022422-20180717-C00068
Figure US10022422-20180717-C00069
Figure US10022422-20180717-C00070
Figure US10022422-20180717-C00071
Figure US10022422-20180717-C00072
Figure US10022422-20180717-C00073
Figure US10022422-20180717-C00074
Figure US10022422-20180717-C00075
Figure US10022422-20180717-C00076
Figure US10022422-20180717-C00077
Figure US10022422-20180717-C00078
Figure US10022422-20180717-C00079
Figure US10022422-20180717-C00080
Figure US10022422-20180717-C00081
Figure US10022422-20180717-C00082
Figure US10022422-20180717-C00083
Figure US10022422-20180717-C00084
Figure US10022422-20180717-C00085
Figure US10022422-20180717-C00086
Figure US10022422-20180717-C00087
Figure US10022422-20180717-C00088
Figure US10022422-20180717-C00089
Figure US10022422-20180717-C00090
Figure US10022422-20180717-C00091
Figure US10022422-20180717-C00092
Figure US10022422-20180717-C00093
Figure US10022422-20180717-C00094
Figure US10022422-20180717-C00095
Figure US10022422-20180717-C00096
Figure US10022422-20180717-C00097
Figure US10022422-20180717-C00098
Figure US10022422-20180717-C00099
Figure US10022422-20180717-C00100
Figure US10022422-20180717-C00101
Figure US10022422-20180717-C00102
Figure US10022422-20180717-C00103
Figure US10022422-20180717-C00104
Figure US10022422-20180717-C00105
Figure US10022422-20180717-C00106
Figure US10022422-20180717-C00107
Each X and Y in this table, is, for example, independently Cl—, Br— or I—.
Additional methods of forming peptidomimetic macrocycles which are envisioned as suitable to perform the present invention include those disclosed by Mustapa, M. Firouz Mohd et al., J. Org. Chem (2003), 68, pp. 8193-8198; Yang, Bin et al. Bioorg Med. Chem. Left. (2004), 14, pp. 1403-1406; U.S. Pat. No. 5,364,851; U.S. Pat. No. 5,446,128; U.S. Pat. No. 5,824,483; U.S. Pat. No. 6,713,280; and U.S. Pat. No. 7,202,332. In such embodiments, aminoacid precursors are used containing an additional substituent R— at the alpha position. Such aminoacids are incorporated into the macrocycle precursor at the desired positions, which may be at the positions where the crosslinker is substituted or, alternatively, elsewhere in the sequence of the macrocycle precursor. Cyclization of the precursor is then effected according to the indicated method.
In some embodiments, it is desirable to modify the configuration of the resulting peptidomimetic macrocycle. For instance, when a 310 helical configuration is more desirable, additional substitutions or modifications to the macrocycle can be made to induce or bias such conformations, such as substituting 2-aminoisobutyric acid (Aib) for one or more amino acids in the sequence of the invention. See, for example, Boal et. al., J. Am. Chem. Soc. 2007, 129, 6986-6987. In one embodiments, the helical macrocycle of the invention comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more Aib substitutions.
Assays
The properties of the peptidomimetic macrocycles of the invention are assayed, for example, by using the methods described below. In some embodiments, a peptidomimetic macrocycle of the invention has improved biological properties relative to a corresponding polypeptide lacking the substituents described herein.
Assay to Determine Helicity.
In solution, the secondary structure of polypeptides with helical domains will reach a dynamic equilibrium between random coil structures and helical structures, often expressed as a “percent helicity”. Thus, for example, unmodified helical domains may be predominantly random coils in solution, with helical content usually under 25%. Peptidomimetic macrocycles with optimized linkers, on the other hand, possess, for example, a helicity that is at least two-fold greater than that of a corresponding uncrosslinked polypeptide. In some embodiments, macrocycles of the invention will possess a helicity of greater than 50%. To assay the helicity of peptidomimetic macrocyles of the invention, the compounds are dissolved in an aqueous solution (e.g. 50 mM potassium phosphate solution at pH 7, or distilled H2O, to concentrations of 25-50 μM). Circular dichroism (CD) spectra are obtained on a spectropolarimeter (e.g., Jasco J-710) using standard measurement parameters (e.g. temperature, 20° C.; wavelength, 190-260 nm; step resolution, 0.5 nm; speed, 20 nm/sec; accumulations, 10; response, 1 sec; bandwidth, 1 nm; path length, 0.1 cm). The helical content of each peptide is calculated by dividing the mean residue ellipticity (e.g. [Φ]222obs) by the reported value for a model helical decapeptide (Yang et al. (1986), Methods Enzymol. 130:208)).
Assay to Determine Melting Temperature (Tm).
A peptidomimetic macrocycle of the invention comprising a secondary structure such as a helix exhibits, for example, a higher melting temperature than a corresponding uncrosslinked polypeptide. Typically peptidomimetic macrocycles of the invention exhibit Tm of >60° C. representing a highly stable structure in aqueous solutions. To assay the effect of macrocycle formation on melting temperature, peptidomimetic macrocycles or unmodified peptides are dissolved in distilled H2O (e.g. at a final concentration of 50 μM) and the Tm is determined by measuring the change in ellipticity over a temperature range (e.g. 4 to 95° C.) on a spectropolarimeter (e.g., Jasco J-710) using standard parameters (e.g. wavelength 222 nm; step resolution, 0.5 nm; speed, 20 nm/sec; accumulations, 10; response, 1 sec; bandwidth, 1 nm; temperature increase rate: 1° C./min; path length, 0.1 cm).
Protease Resistance Assay.
The amide bond of the peptide backbone is susceptible to hydrolysis by proteases, thereby rendering peptidic compounds vulnerable to rapid degradation in vivo. Peptide helix formation, however, typically buries the amide backbone and therefore may shield it from proteolytic cleavage. The peptidomimetic macrocycles of the present invention may be subjected to in vitro trypsin proteolysis to assess for any change in degradation rate compared to a corresponding uncrosslinked polypeptide. For example, the peptidomimetic macrocycle and a corresponding uncrosslinked polypeptide are incubated with trypsin agarose and the reactions quenched at various time points by centrifugation and subsequent HPLC injection to quantitate the residual substrate by ultraviolet absorption at 280 nm. Briefly, the peptidomimetic macrocycle and peptidomimetic precursor (5 mcg) are incubated with trypsin agarosc (Pierce) (S/E ˜125) for 0, 10, 20, 90, and 180 minutes. Reactions are quenched by tabletop centrifugation at high speed; remaining substrate in the isolated supernatant is quantified by HPLC-based peak detection at 280 nm. The proteolytic reaction displays first order kinetics and the rate constant, k, is determined from a plot of ln [S] versus time (k=−1×slope).
Ex Vivo Stability Assay.
Peptidomimetic macrocycles with optimized linkers possess, for example, an ex vivo half-life that is at least two-fold greater than that of a corresponding uncrosslinked polypeptide, and possess an ex vivo half-life of 12 hours or more. For ex vivo serum stability studies, a variety of assays may be used. For example, a peptidomimetic macrocycle and a corresponding uncrosslinked polypeptide (2 meg) are incubated with fresh mouse, rat and/or human serum (2 mL) at 37° C. for 0, 1, 2, 4, 8, and 24 hours. To determine the level of intact compound, the following procedure may be used: The samples are extracted by transferring 100 μl of sera to 2 ml centrifuge tubes followed by the addition of 10 μL of 50% formic acid and 500 μL acetonitrile and centrifugation at 14,000 RPM for 10 min at 4±2° C. The supernatants are then transferred to fresh 2 ml tubes and evaporated on Turbovap under N2<10 psi, 37° C. The samples are reconstituted in 100 μL, of 50:50 acetonitrile:water and submitted to LC-MS/MS analysis.
In Vitro Binding Assays.
To assess the binding and affinity of peptidomimetic macrocycles and peptidomimetic precursors to acceptor proteins, a fluorescence polarization assay (FPA) issued, for example. The FPA technique measures the molecular orientation and mobility using polarized light and fluorescent tracer. When excited with polarized light, fluorescent tracers (e.g., FITC) attached to molecules with high apparent molecular weights (e.g. FITC-labeled peptides bound to a large protein) emit higher levels of polarized fluorescence due to their slower rates of rotation as compared to fluorescent tracers attached to smaller molecules (e.g. FITC-labeled peptides that are free in solution).
For example, fluoresceinated peptidomimetic macrocycles (25 nM) are incubated with the acceptor protein (25-1000 nM) in binding buffer (140 mM NaCl, 50 mM Tris-HCL, pH 7.4) for 30 minutes at room temperature. Binding activity is measured, for example, by fluorescence polarization on a luminescence spectrophotometer (e.g. Perkin-Elmer LS50B). Kd values may be determined by nonlinear regression analysis using, for example, Graphpad Prism software (GraphPad Software, Inc., San Diego, Calif.). A peptidomimetic macrocycle of the invention shows, in some instances, similar or lower Kd than a corresponding uncrosslinked polypeptide.
In Vitro Displacement Assays to Characterize Antagonists of Peptide-Protein Interactions.
To assess the binding and affinity of compounds that antagonize the interaction between a peptide and an acceptor protein, a fluorescence polarization assay (FPA) utilizing a fluoresceinated peptidomimetic macrocycle derived from a peptidomimetic precursor sequence is used, for example. The FPA technique measures the molecular orientation and mobility using polarized light and fluorescent tracer. When excited with polarized light, fluorescent tracers (e.g., FITC) attached to molecules with high apparent molecular weights (e.g. FITC-labeled peptides bound to a large protein) emit higher levels of polarized fluorescence due to their slower rates of rotation as compared to fluorescent tracers attached to smaller molecules (e.g. FITC-labeled peptides that are free in solution). A compound that antagonizes the interaction between the fluoresceinated peptidomimetic macrocycle and an acceptor protein will be detected in a competitive binding FPA experiment.
For example, putative antagonist compounds (1 nM to 1 mM) and a fluoresceinated peptidomimetic macrocycle (25 nM) are incubated with the acceptor protein (50 nM) in binding buffer (140 mM NaCl, 50 mM Tris-HCL, pH 7.4) for 30 minutes at room temperature. Antagonist binding activity is measured, for example, by fluorescence polarization on a luminescence spectrophotometer (e.g. Perkin-Elmer LS50B). Kd values may be determined by nonlinear regression analysis using, for example, Graphpad Prism software (GraphPad Software, Inc., San Diego, Calif.).
Any class of molecule, such as small organic molecules, peptides, oligonucleotides or proteins can be examined as putative antagonists in this assay.
Binding Assays in Intact Cells.
It is possible to measure binding of peptides or peptidomimetic macrocycles to their natural acceptors in intact cells by immunoprecipitation experiments. For example, intact cells are incubated with fluoresceinated (FITC-labeled) compounds for 4 hrs in the absence of serum, followed by serum replacement and further incubation that ranges from 4-18 hrs. Cells are then pelleted and incubated in lysis buffer (50 mM Tris [pH 7.6], 150 mM NaCl, 1% CHAPS and protease inhibitor cocktail) for 10 minutes at 4° C. Extracts are centrifuged at 14,000 rpm for 15 minutes and supernatants collected and incubated with 10 μl goat anti-FITC antibody for 2 hrs, rotating at 4° C. followed by further 2 hrs incubation at 4° C. with protein A/G Sepharose (50 μl of 50% bead slurry). After quick centrifugation, the pellets are washed in lysis buffer containing increasing salt concentration (e.g., 150, 300, 500 mM). The beads are then re-equilibrated at 150 mM NaCl before addition of SDS-containing sample buffer and boiling. After centrifugation, the supernatants are optionally electrophoresed using 4%-12% gradient Bis-Tris gels followed by transfer into Immobilon-P membranes. After blocking, blots are optionally incubated with an antibody that detects FITC and also with one or more antibodies that detect proteins that bind to the peptidomimetic macrocycle.
Cellular Penetrability Assays.
A peptidomimetic macrocycle is, for example, more cell penetrable compared to a corresponding uncrosslinked macrocycle. Peptidomimetic macrocycles with optimized linkers possess, for example, cell penetrability that is at least two-fold greater than a corresponding uncrosslinked macrocycle, and often 20% or more of the applied peptidomimetic macrocycle will be observed to have penetrated the cell after 4 hours. To measure the cell penetrability of peptidomimetic macrocycles and corresponding uncrosslinked macrocycle, intact cells are incubated with fluoresceinated peptidomimetic macrocycles or corresponding uncrosslinked macrocycle (10 μM) for 4 hrs in serum free media at 37° C., washed twice with media and incubated with trypsin (0.25%) for 10 min at 37° C. The cells are washed again and resuspended in PBS. Cellular fluorescence is analyzed, for example, by using either a FACS Calibur flow cytometer or Cellomics' KineticScan® HCS Reader.
In Vivo Stability Assay.
To investigate the in vivo stability of the peptidomimetic macrocycles, the compounds are, for example, administered to mice and/or rats by IV, IP, PO or inhalation routes at concentrations ranging from 0.1 to 50 mg/kg and blood specimens withdrawn at 0′, 5′, 15′, 30′, 1 hr, 4 hrs, 8 hrs and 24 hours post-injection. Levels of intact compound in 25 μL of fresh scrum are then measured by LC-MS/MS as above.
In Vitro Testing for Inhibition of Influenza Replication
This influenza antiviral evaluation assay examines the effects of compounds at designated dose-response concentrations. See also Noah, J. W., W. Severson, D. L. Noah, L. Rasmussen, E. L. White, and C. B. Jonsson, Antiviral Res, 2007. 73(1): p. 50-9. Malin Darby canine kidney (MDCK) cells are used in the assay to test the efficacy of the compounds in preventing the cytopathic effect (CPE) induced by influenza infection. Either Ribavirin or Tamiflu is included in each run as a positive control compound. Subconfluent cultures of MDCK cells are plated into 96-well plates for the analysis of cell viability (cytotoxicity) and antiviral activity (CPE). Drugs are added to the cells 24 hours later. At a designated time, the CPE wells also receive 100 tissue culture infectious doses (100 TCID50s) of titered influenza virus. 72 hours later the cell viability is determined. The effective compound concentrations which reduce viral-induced CPE by 25% (IC25), 50% (IC50), and 90% (IC90) are calculated by regression analysis with semi-log curve fitting. Cell viability is assessed using CellTiter-Glo (Promega). The toxic concentration of drug that reduces cell numbers by 50% and 90% (IC50 and TC90, respectively) are calculated as well. Selectivity (therapeutic) indices (SI=TC/IC) are also calculated.
In Vivo Testing for Inhibition of Influenza Replication
In vivo testing of compounds of the invention can be performed, including testing on mammals such as rats or ferrets. Because ferrets (Mustela putorius furo) are naturally susceptible to infection with human influenza A and B viruses and their disease resembles that of human influenza, these animals have been widely used as a model for influenza virus pathogenesis and immunity studies. Sec Sidwell, R. W. and D. F. Smec, Antiviral Res, 2000. 48(1): p. 1-16; and Colacino, J. M., D. C. DeLong, J. R. Nelson, W. A. Spitzer, J. Tang, F. Victor, and C. Y. Wu, Antimicrob Agents Chemother, 1990. 34(11): p. 2156-63. Ferrets are also the model of choice for the study of avian influenza virus H5N1 pathogenesis in mammals. See also Zitzow, L. A., T. Rowe, T. Morken, W.-J. Shieh, S. Zaki, and J. M. Katz, Pathogenesis of Avian Influenza A (H5N1) Viruses in Ferrets. 2002. p. 4420-4429. The activities of the PB1 Stapled Peptides can be compared to Ribavirin or Oseltamivir as a positive control.
Briefly, young adult male or female ferrets (five ferrets for each treatment group) that are serologically negative by hemagglutination inhibition assay for currently circulating human influenza A or B viruses are quarantined at least 4 days prior to infection in a BSL-3+ animal holding area, where they are housed in cages contained in bioclean portable laminar flow clean room enclosures (Lab Products, Seaford, Del.). Prior to infection, baseline temperatures are measured twice daily for at least 3 days. Ferrets are anesthetized with ketamine (25 mg/kg), xylazine (2 mg/kg), and atropine (0.05 mg/kg) by the intramuscular route and infected intranasally (i.n.) with virus/ml in phosphate-buffered saline (PBS) delivered to the nostrils. Control animals are mock-infected with an equivalent dilution (1:30) of noninfectious allantoic fluid. Stapled Peptides are administered i.v. or i.p. one hour after virus infection.
Temperatures are measured twice daily using either a rectal thermometer or a subcutaneous implantable temperature transponder (BioMedic Data Systems, Inc., Seaford, Del.) with pre-infection values averaged to obtain a baseline temperature for each ferret. The change in temperature (in degrees Celsius) is calculated at each time point for each animal. Clinical signs of sneezing (before anesthesia), inappetence, dyspnea, and level of activity are assessed. A scoring system is also used to assess the activity level, and based on the daily scores for each animal in a group a relative inactivity index will be calculated. Rectal temperature and activity scores are used to assess the severity of influenza infection and the ability of Stapled Peptides to prevent flu symptoms.
Assaying Inhibition of Viral Polymerase Complex Assembly and Activity.
The technique of Bimolecular Fluorescence Complementation (“BiFC”) may be used to assay the compounds of the invention. In this technique, N- and C-terminal fragments of fluorescent proteins (e.g. GFP or its derivatives) are fused to interacting proteins. The two non-functional halves of the fluorophore, following the expression in cells, are brought into close proximity as a result of the specific protein interactions, which initiates folding of the fragments into an active protein and results in a detectable fluorescent signal at the site of the protein-protein complex. Thus, through BiFC, the specific interaction between PB1 and PA subunits can be visualized, quantified and localized within live cells. By disrupting PB1-PA interaction with a compound of the invention, the BiFC signal will be reduced, indicative of the presence of potential inhibitors targeting the assembly of PB1-PA complex. See Hemerka et. al., J. Virol. 2009, 3944-3955.
Pharmaceutical Compositions and Routes of Administration
The peptidomimetic macrocycles of the invention also include pharmaceutically acceptable derivatives or prodrugs thereof. A “pharmaceutically acceptable derivative” means any pharmaceutically acceptable salt, ester, salt of an ester, pro-drug or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention. Particularly favored pharmaceutically acceptable derivatives are those that increase the bioavailability of the compounds of the invention when administered to a mammal (e.g., by increasing absorption into the blood of an orally administered compound) or which increases delivery of the active compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species. Some pharmaceutically acceptable derivatives include a chemical group which increases aqueous solubility or active transport across the gastrointestinal mucosa.
In some embodiments, the peptidomimetic macrocycles of the invention are modified by covalently or non-covalently joining appropriate functional groups to enhance selective biological properties. Such modifications include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism, and alter rate of excretion.
Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acid salts include acetate, adipate, benzoate, benzenesulfonate, butyrate, citrate, digluconate, dodecylsulfate, formate, fumarate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, palmoate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, tosylate and undecanoate. Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N-(alkyl)4 + salts.
For preparing pharmaceutical compositions from the compounds of the present invention, pharmaceutically acceptable carriers include either solid or liquid carriers. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substances, which also acts as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. Details on techniques for formulation and administration are well described in the scientific and patent literature, see, e.g., the latest edition of Remington's Pharmaceutical Sciences, Maack Publishing Co, Easton Pa.
In powders, the carrier is a finely divided solid, which is in a mixture with the finely divided active component. In tablets, the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
Suitable solid excipients are carbohydrate or protein fillers include, but are not limited to sugars, including lactose, sucrose, mannitol, or sorbitol; starch from corn, wheat, rice, potato, or other plants; cellulose such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; and gums including arabic and tragacanth; as well as proteins such as gelatin and collagen. If desired, disintegrating or solubilizing agents are added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid, or a salt thereof, such as sodium alginate.
Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water/propylene glycol solutions. For parenteral injection, liquid preparations can be formulated in solution in aqueous polyethylene glycol solution.
The pharmaceutical preparation is preferably in unit dosage form. In such form the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself; or it can be the appropriate number of any of these in packaged form.
When the compositions of this invention comprise a combination of a peptidomimetic macrocycle and one or more additional therapeutic or prophylactic agents, both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen. In some embodiments, the additional agents are administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those agents are part of a single dosage form, mixed together with the compounds of this invention in a single composition.
Methods of Use
Generally, the invention discloses peptidomimetic macrocycles useful in the treatment of viral disorders. For example, peptidomimetic macrocycles derived from the PB1 helix sequence, or peptidomimetic macrocycles that bind selectively to the PB1 peptide binding site of the PA protein, may selectively inhibit influenza RNA-dependent RNA polymerases. Peptidomimetic macrocycles derived from the PB2 helix sequence, or peptidomimetic macrocycles that bind selectively to the PB2 peptide binding site of the PB1 protein, may selectively inhibit influenza RNA-dependent RNA polymerases. When administered within a therapeutic window after infection, such peptidomimetic macrocycles may reduce the severity or duration of an influenza infection. When administered prophylactically, such peptidomimetic macrocycles may prevent infection by influenza viruses and thereby decrease the spread of influenza and reduce large-scale epidemics.
In one aspect, the present invention provides novel peptidomimetic macrocycles that are useful in competitive binding assays to identify agents which bind to the natural ligand(s) of the proteins or peptides upon which the peptidomimetic macrocycles am modeled. For example, in the PB1/PA system, labeled peptidomimetic macrocycles based on PB1 can be used in a PA binding assay along with small molecules that competitively bind to PA. Competitive binding studies allow for rapid in vitro evaluation and determination of drug candidates specific for the PB1/PA system. Such binding studies may be performed with any of the peptidomimetic macrocycles disclosed herein and their binding partners.
In other aspects, the present invention provides for both prophylactic and therapeutic methods of treating a subject infected with, at risk of, or susceptible to an influenza virus. These methods comprise administering an effective amount of a compound of the invention to a warm blooded animal, including a human. Tn some embodiments, the administration of the compounds of the present invention prevents the proliferation or transmission of an influenza virus.
As used herein, the term “treatment” is defined as the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease or the predisposition toward disease.
In some embodiments, peptidomimetic macrocycles of the invention are used to treat diseases induced by influenza viruses. Like other viruses, the replication of influenza virus involves six phases; transmission, entry, replication, biosynthesis, assembly, and exit. Entry occurs by endocytosis, replication and vRNP assembly takes place in the nucleus, and the virus buds from the plasma membrane. In the infected patient, the virus targets airway epithelial cells.
The methods described herein are also useful for development and/or identification of agents for the treatment of infections caused by viruses such as Abelson leukemia virus, Abelson murine leukemia virus, Abelson's virus, Acute laryngotracheobronchitis virus, Adelaide River virus, Adeno associated virus group, Adenovirus, African horse sickness virus, African swine fever virus, AIDS virus, Aleutian mink disease parvovirus, Alpharetrovirus, Alphavirus, ALV related virus, Amapari virus, Aphthovirus, Aquareovirus, Arbovirus, Arbovirus C, arbovirus group A, arbovirus group B, Arenavirus group, Argentine hemorrhagic fever virus, Argentine hemorrhagic fever virus, Arterivirus, Astrovirus, Ateline herpesvirus group, Aujezky's disease virus, Aura virus, Ausduk disease virus, Australian bat lyssavirus, Aviadenovirus, avian erythroblastosis virus, avian infectious bronchitis virus, avian leukemia virus, avian leukosis virus, avian lymphomatosis virus, avian myeloblastosis virus, avian paramyxovirus, avian pneumoencephalitis virus, avian reticuloendotheliosis virus, avian sarcoma virus, avian type C retrovirus group, Avihepadnavirus, Avipoxvirus, B virus, B19 virus, Babanki virus, baboon herpesvirus, baculovirus, Barmah Forest virus, Bebaru virus, Berrimah virus, Betaretrovirus, Birnavirus, Bittner virus, BK virus, Black Creek Canal virus, bluetongue virus, Bolivian hemorrhagic fever virus, Boma disease virus, border disease of sheep virus, boma virus, bovine alphaherpesvirus 1, bovine alphaherpesvirus 2, bovine coronavirus, bovine ephemeral fever virus, bovine immunodeficiency virus, bovine leukemia virus, bovine leukosis virus, bovine mammillitis virus, bovine papillomavirus, bovine papular stomatitis virus, bovine parvovirus, bovine syncytial virus, bovine type C oncovirus, bovine viral diarrhea virus, Buggy Creek virus, bullet shaped virus group, Bunyamwera virus supergroup, Bunyavirus, Burkitt's lymphoma virus, Bwamba Fever, CA virus, Calicivirus, California encephalitis virus, camelpox virus, canarypox virus, canid herpesvirus, canine coronavirus, canine distemper virus, canine herpesvirus, canine minute virus, canine parvovirus, Cano Delgadito virus, caprine arthritis virus, caprine encephalitis virus, Caprine Herpes Virus, Capripox virus, Cardiovirus, caviid herpesvirus 1, Cercopithecid herpesvirus 1, cercopithecine herpesvirus 1, Cercopithecine herpesvirus 2, Chandipura virus, Changuinola virus, channel catfish virus, Charkville virus, chickenpox virus, Chikungunya virus, chimpanzee herpesvirus, chub reovirus, chum salmon virus, Cocal virus, Coho salmon reovirus, coital exanthema virus, Colorado tick fever virus, Coltivirus, Columbia SK virus, common cold virus, contagious ecthyma virus, contagious pustular dermatitis virus, Coronavirus, Corriparta virus, coryza virus, cowpox virus, coxsackie virus, CPV (cytoplasmic polyhedrosis virus), cricket paralysis virus, Crimean-Congo hemorrhagic fever virus, croup associated virus, Cryptovirus, Cypovirus, Cytomegalovirus, cytomegalovirus group, cytoplasmic polyhedrosis virus, deer papillomavirus, deltaretrovirus, dengue virus, Densovirus, Dependovirus, Dhori virus, diploma virus, Drosophila C virus, duck hepatitis B virus, duck hepatitis virus 1, duck hepatitis virus 2, duovirus, Duvenhage virus, Deformed wing virus DWV, eastern equine encephalitis virus, eastern equine encephalomyelitis virus, ER virus, Ebola virus, Ebola-like virus, echo virus, echovirus, echovirus 10, echovirus 28, echovirus 9, ectromelia virus, EEE virus, EIA virus, EIA virus, encephalitis virus, encephalomyocarditis group virus, encephalomyocarditis virus, Enterovirus, enzyme elevating virus, enzyme elevating virus (LDH), epidemic hemorrhagic fever virus, epizootic hemorrhagic disease virus, Epstein-Barr virus, equid alphaherpesvirus 1, equid alphaherpesvirus 4, equid herpesvirus 2, equine abortion virus, equine arteritis virus, equine encephalosis virus, equine infectious anemia virus, equine morbillivirus, equine rhinopneumonitis virus, equine rhinovirus, Eubenangu virus, European elk papillomavirus, European swine fever virus, Everglades virus, Eyach virus, felid herpesvirus 1, feline calicivirus, feline fibrosarcoma virus, feline herpesvirus, feline immunodeficiency virus, feline infectious peritonitis virus, feline leukemia/sarcoma virus, feline leukemia virus, feline panleukopenia virus, feline parvovirus, feline sarcoma virus, feline syncytial virus, Filovirus, Flanders virus, Flavivirus, foot and mouth disease virus, Fort Morgan virus, Four Corners hantavirus, fowl adenovirus 1, fowlpox virus, Friend virus, Gammaretrovirus, GB hepatitis virus, GB virus, German measles virus, Getah virus, gibbon ape leukemia virus, glandular fever virus, goatpox virus, golden shinner virus, Gonometa virus, goose parvovirus, granulosis virus, Gross' virus, ground squirrel hepatitis B virus, group A arbovirus, Guanarito virus, guinea pig cytomegalovirus, guinea pig type C virus, Hantaan virus, Hantavirus, hard clam reovirus, hare fibroma virus, HCMV (human cytomegalovirus), hemadsorption virus 2, hemagglutinating virus of Japan, hemorrhagic fever virus, hendra virus, Henipaviruses, Hepadnavirus, hepatitis A virus, hepatitis B virus group, hepatitis C virus, hepatitis D virus, hepatitis delta virus, hepatitis E virus, hepatitis F virus, hepatitis G virus, hepatitis nonA nonB virus, hepatitis virus, hepatitis virus (nonhuman), hepatoencephalomyelitis reovirus 3, Hepatovirus, heron hepatitis B virus, herpes B virus, herpes simplex virus, herpes simplex virus 1, herpes simplex virus 2, herpesvirus, herpesvirus 7, Herpesvirus ateles, Herpesvirus hominis, Herpesvirus infection, Herpesvirus saimiri, Herpesvirus suis, Herpesvirus varicellae, Highlands J virus, Hirame rhabdovirus, hog cholera virus, human adenovirus 2, human alphaherpesvirus 1, human alphaherpesvirus 2, human alphaherpesvinis 3, human B lymphotropic virus, human betaherpesvirus 5, human coronavirus, human cytomegalovirus group, human foamy virus, human gammaherpesvirus 4, human gammaherpesvirus 6, human hepatitis A virus, human herpesvirus 1 group, human herpesvirus 2 group, human herpesvirus 3 group, human herpesvirus 4 group, human herpesvirus 6, human herpesvirus 8, human immunodeficiency virus, human immunodeficiency virus 1, human immunodeficiency virus 2, human papillomavirus, human T cell leukemia virus, human T cell leukemia virus I, human T cell leukemia virus II, human T cell leukemia virus DI, human T cell lymphoma virus I, human T cell lymphoma virus II, human T cell lymphotropic virus type 1, human T cell lymphotropic virus type 2, human T lymphotropic virus I, human T lymphotropic virus II, human T lymphotropic virus III, Ichnovirus, infantile gastroenteritis virus, infectious bovine rhinotracheitis virus, infectious haematopoietic necrosis virus, infectious pancreatic necrosis virus, influenza virus A, influenza virus B, influenza virus C, influenza virus D, influenza virus pr8, insect iridescent virus, insect virus, iridovirus, Japanese B virus, Japanese encephalitis virus, JC virus, Junin virus, Kaposi's sarcoma-associated herpesvirus, Kemerovo virus, Kilham's rat virus, Klamath virus, Kolongo virus, Korean hemorrhagic fever virus, kumba virus, Kysanur forest disease virus, Kyzylagach virus, La Crosse virus, lactic dehydrogenase elevating virus, lactic dehydrogenase virus, Lagos bat virus, Langur virus, lapine parvovirus, Lassa fever virus, Lassa virus, latent rat virus, LCM virus, Leaky virus, Lentivirus, Leporipoxvirus, leukemia virus, leukovirus, lumpy skin disease virus, lymphadenopathy associated virus, Lymphocryptovirus, lymphocytic choriomeningitis virus, lymphoproliferative virus group, Machupo virus, mad itch virus, mammalian type B oncovirus group, mammalian type B retroviruses, mammalian type C retrovirus group, mammalian type D retroviruses, mammary tumor virus, Mapuera virus, Marburg virus, Marburg-like virus, Mason Pfizer monkey virus, Mastadenovirus, Mayaro virus, ME virus, measles virus, Menangle virus, Mengo virus, Mengovirus, Middelburg virus, milkers nodule virus, mink enteritis virus, minute virus of mice, MLV related virus, MM virus, Mokola virus, Molluscipoxvirus, Molluscum contagiosum virus, monkey B virus, monkeypox virus, Mononegavirales, Morbillivirus, Mount Elgon bat virus, mouse cytomegalovirus, mouse encephalomyelitis virus, mouse hepatitis virus, mouse K virus, mouse leukemia virus, mouse mammary tumor virus, mouse minute virus, mouse pneumonia virus, mouse poliomyelitis virus, mouse polyomavirus, mouse sarcoma virus, mouscpox virus, Mozambique virus, Mucambo virus, mucosal disease virus, mumps virus, murid betaherpesvirus 1, murid eytomegalovirus 2, murine cytomegalovirus group, murine encephalomyelitis virus, murine hepatitis virus, murine leukemia virus, murine nodule inducing virus, murine polyomavirus, murine sarcoma virus, Muromegalovirus, Murray Valley encephalitis virus, myxoma virus, Myxovirus, Myxovirus multiforme, Myxovirus parotitidis, Nairobi sheep disease virus, Nairovirus, Nanimavirus, Nariva virus, Ndumo virus, Neethling virus, Nelson Bay virus, neurotropic virus, New World Arenavirus, newborn pneumonitis virus, Newcastle disease virus, Nipah virus, noncytopathogenic virus, Norwalk virus, nuclear polyhedrosis virus (NPV), nipple neck virus, O'nyong'nyong virus, Ockelbo virus, oncogenic virus, oncogenic viruslike particle, oncomavirus, Orbivirus, Orf virus, Oropouche virus, Orthohepadnavirus, Orthomyxovirus, Orthopoxvirus, Orthoreovirus, Orungo, ovine papillomavirus, ovine catarrhal fever virus, owl monkey herpesvirus, Palyam virus, Papillomavirus, Papillomavirus sylvilagi, Papovavirus, parainfluenza virus, parainfluenza virus type 1, parainfluenza virus type 2, parainfluenza virus type 3, parainfluenza virus type 4, Paramyxovirus, Parapoxvirus, paravaccinia virus, Parvovirus, Parvovirus B19, parvovirus group, Pestivirus, Phlebovirus, phocine distemper virus, Picodnavirus, Picornavirus, pig cytomegalovirus-pigconpox virus, Piry virus, Pixuna virus, pneumonia virus of mice, Pneumovirus, poliomyelitis virus, poliovirus, Polydnavirus, polyhedral virus, polyoma virus, Polyomavirus, Polyomavirus bovis, Polyomavirus cercopitheci, Polyomavirus hominis 2, Polyomavirus maccacac 1, Polyomavirus muris 1, Polyomavirus muris 2, Polyomavirus papionis 1, Polyomavirus papionis 2, Polyomavirus sylvilagi, Pongine herpesvirus 1, porcine epidemic diarrhea virus, porcine hemagglutinating encephalomyelitis virus, porcine parvovirus, porcine transmissible gastroenteritis virus, porcine type C virus, pox virus, poxvirus, poxvirus variolac, Prospect Hill virus, Provirus, pseudocowpox virus, pseudorabies virus, psittacinepox virus, quailpox virus, rabbit fibroma virus, rabbit kidney vaculolating virus, rabbit papillomavirus, rabies virus, raccoon parvovirus, raccoonpox virus, Ranikhet virus, rat cytomegalovirus, rat parvovirus, rat virus, Rauscher's virus, recombinant vaccinia virus, recombinant virus, reovirus, reovirus 1, reovirus 2, reovirus 3, reptilian type C virus, respiratory infection virus, respiratory syncytial virus, respiratory virus, reticuloendotheliosis virus, Rhabdovirus, Rhabdovirus carpia, Rhadinovirus, Rhinovirus, Rhizidiovirus, Rift Valley fever virus, Riley's virus, rinderpest virus, RNA tumor virus, Ross River virus, Rotavirus, rougeole virus, Rous sarcoma virus, rubella virus, rubeola virus, Rubivirus, Russian autumn encephalitis virus, SA 11 simian virus, SA2 virus, Sabia virus, Sagiyama virus, Saimirine herpesvirus 1, salivary gland virus, sandfly fever virus group, Sandjimba virus, SARS virus, SDAV (sialodacryoadenitis virus), sealpox virus, Semliki Forest Virus, Seoul virus, sheeppox virus, Shope fibroma virus, Shope papilloma virus, simian foamy virus, simian hepatitis A virus, simian human immunodeficiency virus, simian immunodeficiency virus, simian parainfluenza virus, simian T cell lymphotrophic virus, simian virus, simian virus 40, Simplexvirus, Sin Nombre virus, Sindhis virus, smallpox virus, South American hemorrhagic fever viruses, sparrowpox virus, Spumavirus, squirrel fibroma virus, squirrel monkey retrovirus, SSV 1 virus group, STLV (simian T lymphotropic virus) type I, STLV (simian T lymphotropic virus) type II, STLV (simian T lymphotropic virus) type III, stomatitis papulosa virus, submaxillary virus, suid alphaherpesvirus 1, suid herpesvirus 2, Suipoxvirus, swamp fever virus, swinepox virus, Swiss mouse leukemia virus, TAC virus, Tacaribe complex virus, Tacaribe virus, Tanapox virus, Taterapox virus, Tench reovirus, Theiler's encephalomyelitis virus, Theiler's virus, Thogoto virus, Thottapalayam virus, Tick borne encephalitis virus, Tioman virus, Togavirus, Torovirus, tumor virus, Tupaia virus, turkey rhinotracheitis virus, turkeypox virus, type C retroviruses, type D oncovirus, type 17 retrovirus group, ulcerative disease rhabdovirus, Una virus, Uukuniemi virus group, vaccinia virus, vacuolating virus, varicella zoster virus, Varicellovirus, Varicola virus, variola major virus, variola virus, Vasin Gishu disease virus, VEE virus, Venezuelan equine encephalitis virus, Venezuelan equine encephalomyelitis virus, Venezuelan hemorrhagic fever virus, vesicular stomatitis virus, Vesiculovirus, Vilyuisk virus, viper retrovirus, viral haemorrhagic septicemia virus, Visna Maedi virus, Visna virus, volepox virus, VSV (vesicular stomatitis virus), Wallal virus, Warrego virus, wart virus, WEE virus, West Nile virus, western equine encephalitis virus, western equine encephalomyelitis virus, Whataroa virus, Winter Vomiting Virus, woodchuck hepatitis B virus, woolly monkey sarcoma virus, wound tumor virus, WRSV virus, Yaba monkey tumor virus, Yaba virus, Yatapoxvirus, yellow fever virus, and the Yug Bogdanovac virus. In one embodiment an infectome will be produced for each virus that includes an inventory of the host cellular genes involved in virus infection during a specific phase of viral infection, such cellular entry or the replication cycle.
For some viruses a great deal of progress has been made in the elucidation of the steps involved during infection of host cells, and any of these steps may be targeted using peptidomimetic macrocycles of the invention. For example, experiments initiated in the early 1980s showed that influenza virus follows a stepwise, endocytic entry program with elements shared with other viruses such as alpha- and rhabdoviruses (Marsh and Helenius 1989; Whittaker 2006). The steps include: 1) Initial attachment to sialic acid containing glycoconjugates receptors on the cell surface; 2) signaling induced by the virus particle; 3) endocytosis by clathrin-dependent and clathrin-independent cellular mechanism; 4) acid-induced, hemaglutinin (HA)-mediated penetration from late endosomes; 5) acid-activated, M2 and matrix protein (M1) dependent uncoating of the capsid; and, 6) intra-cytosolic transport and nuclear import of vRNPs. These steps depend on assistance from the host cell in the form of sorting receptors, vesicle formation machinery, kinase-mediated regulation, organelle acidification, and, most likely, activities of the cytoskeleton.
Influenza attachment to the cells surface occurs via binding of the HA1 subunit to cell surface glycoproteins and glycolipids that carry oligosaccharide moieties with terminal sialic acid residues (Skehel and Wiley 2000). The linkage by which the sialic acid is connected to the next saccharide contributes to species specificity. Avian strains including H5N1 prefer an a-(2,3)-link and human strains a-(2,6)-link (Matrosovich 2006). In epithelial cells, binding occurs preferentially to microvilli on the apical surface, and endocytosis occurs at base of these extensions (Matlin 1982). Whether receptor binding induces signals that prepare the cell for the invasion is not yet known, but it is likely because activation of protein kinase C and synthesis of phopshatidylinositol-3-phosphate (PI3P) are required for efficient entry (Sieczkarski et al. 2003; Whittaker 2006).
Endocytic internalization occurs within a few minutes after binding (Matlin 1982; Yoshimura and Ohnishi 1984). In tissue culture cells influenza virus makes use of three different types of cellular processes; 1) preexisting clathrin coated pits, 2) virus-induced clathrin coated pits, and 3) endocytosis in vesicles without visible coat (Matlin 1982; Sieczkarski and Whittaker 2002; Rust et al. 2004). Video microscopy using fluorescent viruses showed the virus particles undergoing actin-mediated rapid motion in the cell periphery followed by minus end-directed, microtubule-mediated transport to the perinuclear area of the cell. Live cell imaging indicated that the virus particles first entered a subpopulation of mobile, peripheral early endosomes that carry them deeper into the cytoplasm before penetration takes place (Lakadamyali et al. 2003; Rust et al. 2004). The endocytic process is regulated by protein and lipid kinases, the proteasome, as well as by Rabs and ubiquitin-dependent sorting factors (Khor et al. 2003; Whittaker 2006).
The membrane penetration step is mediated by low pH-mediated activation of the trimeric, metastable HA, and the conversion of this Type I viral fusion protein to a membrane fusion competent conformation (Maeda et al. 1981; White et al. 1982). This occurs about 16 min after internalization, and the pH threshold varies between strains in the 5.0-5.6 range. The target membrane is the limiting membrane of intermediate or late endosomes. The mechanism of fusion has been extensively studied (Kielian and Rey 2006). Further it was observed that fusion itself does not seem to require any host cell components except a lipid bilayer membrane and a functional acidification system (Maeda et al. 1981; White et al. 1982). The penetration step is inhibited by agents such as lysosomotropic weak bases, carboxylic ionophores, and proton pump inhibitors (Malin 1982; Whittaker 2006).
To allow nuclear import of the incoming vRNPs, the capsid has to be disassembled. This step involves acidification of the viral interior through the amantadine-sensitive M2-channels causes dissociation of M1 from the vRNPs (Bukrinskaya et al. 1982; Martin and Helenius 1991; Pinto et al. 1992). Transport of the individual vRNPs to the nuclear pore complexes and transfer into the nucleus depends on cellular nuclear transport receptors (O'Neill et al. 1995; Cros et al. 2005). Replication of the viral RNAs (synthesis of positive and negative strands), and transcription occurs in complexes tightly associated with the chromatin in the nucleus. It is evident that, although many of the steps are catalyzed by the viral polymerase, cellular factors are involved including RNA polymerase activating factors, a chaperone HSP90, hCLE, and a human splicing factor UAP56. Viral gene expression is subject to complex cellular control at the transcriptional level, a control system dependent on cellular kinases (Whittaker 2006).
The final assembly of an influenza particle occurs during a budding process at the plasma membrane. In epithelial cells, budding occurs at the apical membrane domain only (Rodriguez-Boulan 1983). First, the progeny vRNPs are transported within the nucleoplasm to the nuclear envelope, then from the nucleus to the cytoplasm, and finally they accumulate in the cell periphery. Exit from the nucleus is dependent on viral protein NEP and M1, and a variety of cellular proteins including CRM1 (a nuclear export receptor), caspases, and possibly some nuclear protein chaperones. Phosphorylation plays a role in nuclear export by regulating M1 and NEP synthesis, and also through the MAPK/ERK system (Bui et al. 1996; Ludwig 2006). G protein and protein kinase signaling is involved in influenza virus budding from infected host cells (Hui E. and Nayak D, 2002).
The three membrane proteins of the virus are synthesized, folded and assembled into oligomers in the ER (Doms et al. 1993). They pass through the Golgi complex; undergo maturation through modification of their carbohydrate moieties and proteolytic cleavage. After reaching the plasma membrane they associate with M1 and the vRNPs in a budding process that results in the inclusion of all eight vRNPs and exclusion of most host cell components except lipids.
Influenza infection is associated with activation of several signaling cascades including the MAPK pathway (ERK, JNK, p38 and BMK-1/ERK5), the IkB/NF-kB signaling module, the Raf/MEK/ERK cascade, and programmed cell death (Ludwig 2006). These result in a variety of effects that limit the progress of infection such as transcriptional activation of IFNb, apoptotic cell death, and a block in virus escape of from late endosomes (Ludwig 2006).
Example 1
FIGS. 1 and 2 show a possible binding mode of the PB1 helix-derived sequence MDVNPTLLFLKVPAQ (SEQ ID NO: 1). A peptidomimetic macrocycle of the invention is prepared starting with the corresponding uncrosslinked polypeptide sequence MDVNPTLLFLKVPAQ (SEQ ID NO: 1) and replacing the 7th and 10th amino acids with an alpha, alpha-disubstituted amino acid (e.g. the S5 olefin amino acid). An olefin metathesis reaction is performed resulting in a peptidomimetic macrocycle comprising an i to i+3 crosslink as shown in FIG. 2 b.
Example 2
Peptidomimetic macrocycles were synthesized, purified and analyzed as previously described (Walensky et al (2004) Science 305:1466-70; Walensky et al (2006) Mol Cell 24:199-210; Bernal et al (2007) J. Am Chem Soc. 9129, 2456-2457) and as indicated below. The macrocycles used in this study are shown in Table 5. The corresponding uncrosslinked polypeptides represent the natural counterparts of the peptidomimetic macrocycles of the invention.
Alpha,alpha-disubstituted non-natural amino acids containing olefinic side chains were synthesized according to Williams et al. (1991) J. Am. Chem. Soc. 113:9276; Schafmeister et al. (2000) J. Am. Chem Soc. 122:5891 and Verdine et al PCT WO 2008/121767. Peptidomimetic macrocycles were designed by replacing two or more naturally occurring amino acids with the corresponding synthetic amino acids. Substitutions were made at the i and i+3, i and i+4, i and i+6, and i and i+7 positions. Macrocycles were generated by solid phase peptide synthesis followed by olefin metathesis-based crosslinking of the synthetic amino acids via their olefin-containing side chains.
In the sequences shown, the following abbreviations are used: “Nle” represents norleucine, “Aib” represents 2-aminoisobutyric acid, “Ac” represents acetyl, and “Pr” represents propionyl. Amino acids represented as “$” are alpha-Me S5-pentenyl-alanine olefin amino acids connected by an all-carbon i to i+4 crosslinker comprising one double bond. Amino acids represented as “$r5” are alpha-Me R5-pentenyl-alanine olefin amino acids connected by an all-carbon i to i+4 crosslinker comprising one double bond. Amino acids represented as “$s8” are alpha-Me S8-octenyl-alanine olefin amino acids connected by an all-carbon i to i+7 crosslinker comprising one double bond. Amino acids represented as “$r8” are alpha-Me R8-octenyl-alanine olefin amino acids connected by an all-carbon i to i+7 crosslinker comprising one double bond. “Ahx” represents an aminocyclohexyl linker. The crosslinkers are linear all-carbon crosslinker comprising eight or eleven carbon atoms between the alpha carbons of each amino acid. Amino acids represented as “$/” are alpha-Me S5-pentenyl-alanine olefin amino acids that are not connected by any crosslinker. Amino acids represented as “$/r5” are alpha-Me R5-pentenyl-alanine olefin amino acids that are not connected by any crosslinker. Amino acids represented as “$/s8” are alpha-Me S8-octenyl-alanine olefin amino acids that are not connected by any crosslinker. Amino acids represented as “$/r8” are alpha-Me R8-octenyl-alanine olefin amino acids that are not connected by any crosslinker.
The non-natural amino acids (R and S enantiomers of the 5-carbon olefinic amino acid and the S enantiomer of the 8-carbon olefinic amino acid) were characterized by nuclear magnetic resonance (NMR) spectroscopy (Varian Mercury 400) and mass spectrometry (Micromass LCT). Peptide synthesis was performed either manually or on an automated peptide synthesizer (Applied Biosystems, model 433A), using solid phase conditions, rink amide AM resin (Novabiochem), and Fmoc main-chain protecting group chemistry. For the coupling of natural Fmoc-protected amino acids (Novabiochem), 10 equivalents of amino acid and a 1:1:2 molar ratio of coupling reagents HBTU/HOBt (Novabiochem)/DIEA were employed. Non-natural amino acids (4 equiv) were coupled with a 1:1:2 molar ratio of HATU (Applied Biosystems)/HOBt/DIEA. Olefin metathesis was performed in the solid phase using 10 mM Grubbs catalyst (Blackewell et al. 1994 supra) (Strem Chemicals) dissolved in degassed dichloromethane and reacted for 2 hours at room temperature. Isolation of metathesized compounds was achieved by trifluoroacetic acid-mediated deprotection and cleavage, ether precipitation to yield the crude product, and high performance liquid chromatography (HPLC) (Varian ProStar) on a reverse phase C18 column (Varian) to yield the pure compounds. Chemical composition of the pure products was confirmed by LC/MS mass spectrometry (Micromass LCT interfaced with Agilent 1100 HPLC system) and amino acid analysis (Applied Biosystems, model 420A).
The synthesized peptides include a norleucine replacement for methionine to avoid issues with unwanted thioether oxidation. In several peptides, the proline residue is replaced with a 2-aminoisobutyric acid residue (Aib) to increase helicity, and the effect of Glu-to-Arg substitution on cell penetrability was also explored. The N-termini of the synthetic peptides were acetylated, while the C-termini were amidated. Table 5 shows a list of peptidomimetic macrocycles of the invention prepared.
TABLE 5
PB1 Peptidomimetic macrocycles of the invention.
Macro- SEQ Found
cycle ID NO: Sequence EMW M + 2H M + 2H
SP-1 66 Ac-NleDVNPTLLFLKVPAQ-NH2 1707.99 854.995 854.86
SP-2 67 Ac-NleDVNAibTLLFLKVAibAQ-NH2 1683.99 842.995 843.35
SP-3 68 Ac-NleDVNPTLLFLKVPAR-NH2 1736.04 869.02 869.21
SP-4 69 Ac-TLLFLKVPAQ-NH2 1169.72 585.86 585.71
SP-5 70 Ac-TLLF$KVA$Q-NH2 1209.75 605.875 605.75
SP-6 71 Ac-NleDVNAibTLLF$KVA$R-NH2 1764.07 883.035 882.91
SP-7 72 Ac-NleDVNAibTL$FLK$AAR-NH2 1736.04 869.02 869.35
SP-8 73 Ac-NleDVNAibTLLF$KVA$Q-NH2 1736.02 869.01 868.82
SP-9 74 Ac-NleDVNAibTL$FLK$AAQ-NH2 1707.99 854.995 854.86
SP-10 75 Ac-NleDVNPTL$FLK$AAQ-NH2 1719.99 860.995 860.82
SP-11 76 Ac-NleDVNPTLLF$KVA$R-NH2 1776.07 889.035 888.87
SP-12 77 Ac-NleDVNPTL$FLK$AAR-NH2 1748.04 875.02 874.91
SP-13 78 Ac-NleDVNPT$r8LFLKV$AQ-NH2 1790.07 896.035 895.88
SP-14 79 Ac-NleDVNAibT$r8LFLKVA$Q-NH2 1778.07 890.035 890.22
SP-15 80 Ac-NlcDVNAibT$r8LFLKVA$R-NH2 1806.11 904.055 904.25
SP-16 81 Ac-NleDVNPT$r8LFLKVA$Q-NH2 1790.07 896.035 896.24
SP-17 82 Ac-NleDVNPT$r8LFLKVA$R-NH2 1818.11 910.055 910.26
SP-18 83 Ac-NleDVNATLLF$KVA$R-NH2 1750.05 876.025 876.19
SP-19 84 Ac-NleDVNATL$FLK$AAR-NH2 1722.02 862.01 862.23
SP-20 85 Ac-NleDVNATLLF$KVA$Q-NH2 1722.01 862.005 862.23
SP-21 86 Ac-NleDVNATL$FLK$AAQ-NH2 1693.98 847.99 848.21
SP-22 87 Ac-TL$FLK$AAQ-NH2 1181.72 591.86 592.07
SP-23 88 FITC-AhxNleDVNAibTLLF$KVA$Q-NH2 2196.13 1099.065 1099.28
SP-24 89 5-FAM-AhxNleDVNAibTLLFLKVAibAQ-NH2 2113.11 1057.555 1057.85
SP-25 90 5-FAM-AhxNleDVNAibTL$FLK$AAQ-NH2 2137.11 1069.555 1069.82
SP-26 91 5-FAM-AhxNleDVNPTL$FLK$AAQ-NH2 2149.11 1075.555 1075.71
SP-27 92 5-FAM-AhxNleDVNAibT$r8LFLKVA$Q-NH2 2207.19 1104.595 1104.98
SP-28 93 5-FAM-AhxNleDVNPT$r8LFLKVA$Q-NH2 2219.19 1110.595 1110.81
SP-29 94 5-FAM-AhxNleDVNAibT$r5LF$KVAibAR-NH2 2165.16 1083.58 1083.94
SP-30 95 5-FAM-AhxNleDVNAibTLLF$/KVA$/Q-NH2 2193.18 1097.59 1097.92
SP-31 96 Ac-NleDVNAibTLLF$/KVA$/R-NH2 1792.1 897.05 897.3
SP-32 97 Ac-NleDVNPTLLF$/KVA$/R-NH2 1804.1 903.05 903.32
SP-33 98 Ac-NleDVNPTL$/FLK$/AAR-NH2 1776.07 889.035 889.27
SP-34 99 Ac-NleDVNAibTL$/FLK$/AAR-NH2 1764.07 883.035 883.25
SP-35 100 Ac-NleDVNAibT$/r5LF$/KVAibAR-NH2 1764.07 883.035 883.31
SP-36 101 Ac-NleDVNAibTLL$/r5LK$/AAR-NH2 1730.08 866.04 866.29
SP-37 102 Ac-NleDVNAibT$/r8LFLKVA$/R-NH2 1834.15 918.075 918.34
SP-38 103 Ac-NleDVNPT$/r8LFLKVA$/R-NH2 1846.15 924.075 924.36
SP-39 104 Ac-NleDVNPT$/r8LFLKVA$/Q-NH2 1818.1 910.05 910.31
SP-40 105 Ac-NleDVNAT$/r8LFLKVA$/Q-NH2 1792.09 897.045 897.3
SP-41 106 Ac-NleDVNAibT$r5LF$KVAibAR-NH2 1736.04 869.02 869.21
SP-42 107 Ac-NleDVNAibTLL$r5LK$AAR-NH2 1702.05 852.025 852.25
SP-43 108 Ac-NleDVNAT$r8LFLKVA$Q-NH2 1764.06 883.03 883.31
SP-44 109 5-FAM-AhxNleDVNAibTLLF$KVA$Q-NH2 2165.15 1083.575 1083.81
SP-45 110 Ac-NleDVNP$LLF$KVAibAR-NH2 1760.07 881.035 881.31
SP-46 111 Ac-NleDVNPTLL$LKV$AR-NH2 1742.08 872.04 872.31
SP-47 112 Ac-NleDVNAibTLL$LKV$AR-NH2 1730.08 866.04 866.29
SP-48 113 Ac-NleDVN$TLL$LKVAibAQ-NH2 1702.04 852.02 852.3
SP-49 114 Ac-NleDVNP$LLF$KVAibAQ-NH2 1732.03 867.015 867.26
SP-50 115 Ac-NleDVNP$/LLF$/KVAibAR-NH2 1788.1 895.05 895.29
SP-51 116 Ac-NleDVNPTLL$/LKV$/AR-NH2 1770.11 886.055 886.36
SP-52 117 Ac-NleDVNAibTLL$/LKV$/AR-NH2 1758.11 880.055 880.34
SP-53 118 Ac-NleDVN$/TLL$/LKVAibAQ-NH2 1730.07 866.035 866.29
SP-54 119 Ac-NleDVNP$/LLF$/KVAibAQ-NH2 1760.06 881.03 881.31
SP-55 120 Ac-NleDVNAibTLLFLKVAAQ-NH2 1669.98 835.99 836.19
SP-56 121 5-FAM-AhxNleDVNAibTLLF$KVA$R-NH2 2193.19 1097.595 1098.31
SP-57 122 5-FAM-AhxNleDVNPTLLF$KVA$R-NH2 2205.19 1103.595 1104.27
SP-58 123 5-FAM-AhxNleDVNPTL$FLK$AAR-NH2 2177.16 1089.58 1090.15
SP-59 124 5-FAM-AhxNleDVNAibTL$FLK$AAR-NH2 2165.16 1083.58 1084.26
SP-60 125 5-FAM-AhxNleDVNAibTLL$r5LK$AAR-NH2 2131.17 1066.585 1067.29
SP-61 126 5-FAM-AhxNleDVNAibT$r8LFLKVA$R-NH2 2235.23 1118.615 1119.29
SP-62 127 5-FAM-AhxNleDVNPT$r8LFLKVA$R-NH2 2247.23 1124.615 1125.31
SP-63 128 5-FAM-AhxNleDVNAT$r8LFLKVA$Q-NH2 2193.18 1097.59 1098.25
SP-64 129 5-FAM-AhxNleDVNAibTLLFLKLVAAQ-NH2 2099.1 1050.55 1050.78
Example 3
The ex vivo serum stability of several PB1 peptidomimetic macrocycles was tested by incubating them at 5000 ng/mL (2 μM at MW=2500) with fresh human serum at 37° C. and taking samples at 0, 0.5, 1, 2, 4, 6 and 24 hours. At each time point the samples were flash-frozen until analysis in duplicate, then extracted by transferring 100 μl of sera to 2 ml centrifuge tubes followed by the addition of 10 μL of 50% formic acid and 500 μL acetonitrile and centrifugation at 14,000 RPM for 10 min at 4±2° C. After protein precipitation, the supernatants were then transferred to fresh 2 ml tubes and evaporated on Turbovap under N2 at <10 psi, 37° C. The samples were reconstituted in 100 μL of 50:50 acetonitrile/water and quantified by LC-MS/MS analysis. The response for each compound was normalized to estimate a percent decrease in concentration versus time; the results are shown in FIG. 3.
Example 4
Several PB1 peptidomimetic macrocycles were tested for PK properties in single N dose in rats. The in-life portion of the study was conducted at ViviSource Laboratories (Waltham, Mass.). A single intravenous dose of 3 mg/kg Stapled Peptide formulated in water continuing 5% PEG-400 and 2% Dextrose was administered to a pair of jugular vein-cannulated male Sprague-Dawley rats. The N dose was mostly well-tolerated and animals appeared healthy within the study duration. Blood samples were collected over thirteen sampling times up to 24 hours and the plasma samples were shipped on dry ice to Tandem Bioanalytical Facilities, Inc. (Woburn, Mass.) for the analytical phase of the study.
Quantification in plasma samples was preceded by the preparation of sample extracts by combining 50 μl of ammonium hydroxide (14.5 M ammonia), 1 mL of a 1:1 acetonitrile/methanol solution, and 50 μl of internal standard with 50 μl of each plasma sample. The mixtures were centrifuged to separate liquid supernatant from solid precipitate and supernatants were dried at 40° C. under flowing nitrogen gas. The dried sample extracts were reconstituted in 50 μl of a 1:1 water/methanol solution that contained 0.1% (v/v) trifluoroacetic acid. Plasma sample extracts were analyzed by a liquid chromatography-mass spectrometry method that utilized an API 5000 (Applied Biosystems) instrument operated in positive ionization mode at a temperature of 500° C. using a multiple reaction monitoring mode of detection (MRM). The analytical column for liquid chromatography was a Varian Metasil C18, 50 mm×2 mm and mobile phases A (0.1% formic acid in water) and B (0.1% formic acid in acetonitrile) were pumped at a flow rate of 0.5 ml/min. Quantification in plasma extracts was made by linear regression analysis employing a pure reference standard Stapled Peptide diluted in normal rat plasma to prepare eight calibration standards over the working concentration range of 20-10,000 ng/ml. The calibration standards were extracted in identical fashion as sample extracts and analyzed before and after the sample extracts.
Pharmacokinetic parameters were calculated using a non-compartmental model using the PK Functions add-in for Microsoft Excel. The terminal elimination half-life was calculated as ln(2)/(λz), where the rate constant (λz) was calculated as −1 times the estimated slope of the log-concentration versus time data over 2-12 hr. AUC values (hr*ng/ml) were calculated by statistical moment and linear trapezoidal approximation methods over time points of 0-24 hours and 24 hour concentration values were divided by (λz) was added in order to extrapolate AUMC and AUC values to infinite time. Total body clearance (per kg body weight) was calculated as dose divided by AUC. The volume of distribution at steady state (Vss) was calculated as the product of clearance and mean residence time (MRT=AUC/AUMC). The PK results are shown graphically in FIGS. 4, 5 a-5 f, and a table of determined PK parameters is shown in FIG. 6.
An experiment was also performed to compare different modes of administration. Subcutaneous injection of peptidomimetic macrocycle was performed and compared to intravenous administration. Two groups of two animals each were injected subcutaneously with a 3 mg/kg dose. Plasma was collected at regular time points (e.g. 5, 20 minutes; 1, 2, 4, 8 12, and 48 hours) and the samples were analyzed as indicated above. The results are plotted in FIG. 7.
While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (12)

What is claimed is:
1. A composition comprising:
a) a peptidomimetic macrocycle comprising an amino acid sequence wherein two amino acids in the amino acid sequence are cross-linked by a crosslinker, wherein the crosslinker comprises a 1, 5-disubstituted triazole and does not comprise a 1,4-disubstituted triazole;
b) a Cu(I) species; and
c) a Ru(II) species.
2. The composition of claim 1, wherein the peptidomimetic macrocycle comprises a helix.
3. The composition of claim 2, wherein the helix is an alpha helix.
4. The composition of claim 1, wherein the peptidomimetic macrocycle comprises a beta turn.
5. The composition of claim 1, wherein the peptidomimetic macrocycle comprises an α,α-disubstituted amino acid.
6. The composition of claim 1, wherein the peptidomimetic macrocycle comprises more than one stabilized secondary structure.
7. The composition of claim 1, wherein the cross-linked amino acids are at position i and i+3 of the peptidomimetic macrocycle.
8. The composition of claim 1, wherein the crosslinker spans one turn of a secondary structure of the peptidomimetic macrocycle.
9. The composition of claim 1, wherein the crosslinker spans two turns of a secondary structure of the peptidomimetic macrocycle.
10. The composition of claim 1, wherein the peptidomimetic macrocycle comprises more than two cross-linked amino acids.
11. The composition of claim 1, wherein the 1, 5-disubstituted triazole has a formula
Figure US10022422-20180717-C00108
and the 1, 4-disubstituted triazole has a formula
Figure US10022422-20180717-C00109
wherein L1 and L2 are independently alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, cycloarylene, heterocycloarylene, or [—R4—K—R4-]n, each being optionally substituted with R5;
each R4 is alkylene, alkenylene, alkynylene, heteroalkylene, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene;
each K is O, S, SO, SO2, CO, CO2, or CONR3;
each R5 is independently halogen, alkyl, —OR6, —N(R6)2, —SR6, —SOR6, —SO2R6, —CO2R6, a fluorescent moiety, a radioisotope or a therapeutic agent;
each R6 is independently —H, alkyl, alkenyl, alkynyl, arylalkyl, cycloalkylalkyl, heterocycloalkyl, a fluorescent moiety, a radioisotope or a therapeutic agent; and
n is an integer from 1 to 5.
12. The composition of claim 11, wherein each L1 and L2 is independently an alkylene.
US15/093,373 2009-01-14 2016-04-07 Peptidomimetic macrocycles Expired - Fee Related US10022422B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/093,373 US10022422B2 (en) 2009-01-14 2016-04-07 Peptidomimetic macrocycles
US16/002,977 US20180339014A1 (en) 2009-01-14 2018-06-07 Peptidomimetic macrocycles

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US14470609P 2009-01-14 2009-01-14
PCT/US2010/021091 WO2010083347A2 (en) 2009-01-14 2010-01-14 Peptidomimetic macrocycles
US201113129118A 2011-11-01 2011-11-01
US14/853,894 US20160068573A1 (en) 2009-01-14 2015-09-14 Peptidomimetic macrocycles
US15/093,373 US10022422B2 (en) 2009-01-14 2016-04-07 Peptidomimetic macrocycles

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/853,894 Continuation US20160068573A1 (en) 2009-01-14 2015-09-14 Peptidomimetic macrocycles

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/002,977 Division US20180339014A1 (en) 2009-01-14 2018-06-07 Peptidomimetic macrocycles

Publications (2)

Publication Number Publication Date
US20160250278A1 US20160250278A1 (en) 2016-09-01
US10022422B2 true US10022422B2 (en) 2018-07-17

Family

ID=42340294

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/129,118 Expired - Fee Related US9175047B2 (en) 2009-01-14 2010-01-14 Peptidomimetic macrocycles
US14/853,894 Abandoned US20160068573A1 (en) 2009-01-14 2015-09-14 Peptidomimetic macrocycles
US15/093,335 Abandoned US20160251399A1 (en) 2009-01-14 2016-04-07 Peptidomimetic macrocycles
US15/093,373 Expired - Fee Related US10022422B2 (en) 2009-01-14 2016-04-07 Peptidomimetic macrocycles
US16/002,977 Abandoned US20180339014A1 (en) 2009-01-14 2018-06-07 Peptidomimetic macrocycles

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US13/129,118 Expired - Fee Related US9175047B2 (en) 2009-01-14 2010-01-14 Peptidomimetic macrocycles
US14/853,894 Abandoned US20160068573A1 (en) 2009-01-14 2015-09-14 Peptidomimetic macrocycles
US15/093,335 Abandoned US20160251399A1 (en) 2009-01-14 2016-04-07 Peptidomimetic macrocycles

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/002,977 Abandoned US20180339014A1 (en) 2009-01-14 2018-06-07 Peptidomimetic macrocycles

Country Status (8)

Country Link
US (5) US9175047B2 (en)
EP (1) EP2376100B1 (en)
JP (1) JP2012515172A (en)
CN (1) CN102256615A (en)
AU (1) AU2010204648B2 (en)
BR (1) BRPI1006139A2 (en)
CA (1) CA2744088A1 (en)
WO (1) WO2010083347A2 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10213477B2 (en) 2012-02-15 2019-02-26 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10246491B2 (en) 2013-03-06 2019-04-02 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and use thereof in regulating HIF1alpha
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
US10300109B2 (en) 2009-09-22 2019-05-28 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10308699B2 (en) 2011-10-18 2019-06-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10328117B2 (en) 2006-12-14 2019-06-25 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US10471120B2 (en) 2014-09-24 2019-11-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10669230B2 (en) 2012-11-01 2020-06-02 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US10703780B2 (en) 2010-08-13 2020-07-07 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10905739B2 (en) 2014-09-24 2021-02-02 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
US11091522B2 (en) 2018-07-23 2021-08-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US12059413B2 (en) 2016-11-02 2024-08-13 The Research Foundation For The State University Of New York Methods of inhibiting viruses using compositions targeting TSG101-ubiquitin interaction

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008104000A2 (en) 2007-02-23 2008-08-28 Aileron Therapeutics, Inc. Triazole macrocycle systems
EP2342222B1 (en) 2008-09-22 2018-03-21 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
JP2012509902A (en) 2008-11-24 2012-04-26 エルロン・セラピューティクス・インコーポレイテッド Peptidomimetic macrocycles with improved properties
CA2744088A1 (en) 2009-01-14 2010-07-22 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
AU2010306718A1 (en) * 2009-10-14 2012-05-24 Aileron Therapeutics, Inc. Improved peptidomimetic macrocycles
EP2627662B1 (en) 2010-10-13 2015-09-16 Bristol-Myers Squibb Company Methods for preparing macrocycles and macrocycle stabilized peptides
JP6450192B2 (en) 2012-02-15 2019-01-09 エイルロン セラピューティクス,インコーポレイテッド Triazole-bridged and thioether-bridged peptidomimetic macrocycles
US9644002B2 (en) * 2013-07-25 2017-05-09 Yale University Allosteric modulators of EGFR and constitutively active mutants
ES2927607T3 (en) 2013-09-13 2022-11-08 Scripps Research Inst Modified therapeutic agents and compositions thereof
EP4212180A1 (en) 2013-12-18 2023-07-19 The Scripps Research Institute Modified therapeutic agents, stapled peptide lipid conjugates, and compositions thereof
CA2944651A1 (en) * 2014-04-02 2015-10-08 University Of Rochester Macrocyclic peptidomimetics for alpha-helix mimicry
EP3294318A4 (en) 2015-03-20 2019-04-03 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10059741B2 (en) 2015-07-01 2018-08-28 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
JP2018528217A (en) 2015-09-10 2018-09-27 エルロン・セラピューティクス・インコーポレイテッドAileron Therapeutics,Inc. Peptidomimetic macrocycles as modulators of MCL-1
WO2017205786A1 (en) * 2016-05-27 2017-11-30 Aileron Therapeutics, Inc. Cell permeable peptidomimetic macrocycles
EP3559020B1 (en) * 2016-12-22 2022-09-14 Université de Montpellier New stapled-peptides and uses thereof
EP3724216A1 (en) 2017-12-15 2020-10-21 Dana Farber Cancer Institute, Inc. Stabilized peptide-mediated targeted protein degradation
AU2019218786B2 (en) 2018-02-07 2024-05-02 Dana-Farber Cancer Institute, Inc. Cell-permeable stapled peptide modules for cellular delivery
US20240124529A1 (en) * 2020-03-04 2024-04-18 Dana-Farber Cancer Institute, Inc. ANTIVIRAL STRUCTURALLY-STABILIZED SARS-CoV-2 PEPTIDES AND USES THEREOF
WO2021229238A1 (en) * 2020-05-15 2021-11-18 Bicycletx Limited Anti-infective bicyclic peptide ligands
CN117120459A (en) * 2021-01-08 2023-11-24 拜斯科技术开发有限公司 Antiinfective bicyclic peptide ligands

Citations (185)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4000259A (en) 1975-06-16 1976-12-28 American Home Products Corporation Cyclic dodecapeptide analogs of somatostatin and intermediates
US4191754A (en) 1979-02-28 1980-03-04 Merck & Co., Inc. Bicyclic somatostatin analogs
US4438270A (en) 1977-07-11 1984-03-20 Merrell Toraude Et Compagnie α-Halomethyl derivatives of α-amino acids
US4730006A (en) 1986-01-27 1988-03-08 Merrell Dow Pharmaceuticals Inc. Derivatives of 2,6-diamino-3-haloheptanedioic acid
WO1989009233A1 (en) 1988-03-24 1989-10-05 Terrapin Technologies, Inc. Molecular sticks for controlling protein conformation
EP0467699A2 (en) 1990-07-19 1992-01-22 Merck & Co. Inc. Cyclic HIV principal neutralizing determinant peptides
US5120859A (en) 1989-09-22 1992-06-09 Genentech, Inc. Chimeric amino acid analogues
WO1992013878A2 (en) 1991-02-07 1992-08-20 Board Of Trustees Of The University Of Illinois Conformationally restricted mimetics of beta turns and beta bulges and peptides containing the same
WO1993001203A1 (en) 1991-07-11 1993-01-21 Smithkline Beecham Plc Antifungal amphotericin derivative
US5245009A (en) 1990-03-23 1993-09-14 The Salk Institute For Biological Studies CRF antagonists
WO1993019176A1 (en) 1992-03-23 1993-09-30 Imperial Cancer Research Technology Limited Leucine zippers
WO1994025482A1 (en) 1993-04-23 1994-11-10 Evans Herbert J Polypeptides that include conformation-constraining groups which flank a protein-protein interaction site
US5364851A (en) 1991-06-14 1994-11-15 International Synthecon, Llc Conformationally restricted biologically active peptides, methods for their production and uses thereof
WO1995000534A1 (en) 1993-06-18 1995-01-05 The Board Of Trustees Of The University Of Illinois Alpha-helix mimetics and methods relating thereto
WO1996002642A1 (en) 1994-07-20 1996-02-01 University Of Dundee INTERRUPTION OF BINDING OF MDM2 AND p53 PROTEIN AND THERAPEUTIC APPLICATION THEREOF
EP0729972A1 (en) 1995-02-28 1996-09-04 F. Hoffmann-La Roche Ag Peptide derivatives of tetrahydronaphthalene
WO1996028449A1 (en) 1995-03-14 1996-09-19 Hoffman/Barrett, L.L.C. Heteroatom-functionalized porphyrazines and multimetallic complexes and polymers derived therefrom
WO1996034878A1 (en) 1995-05-04 1996-11-07 The Scripps Research Institute Synthesis of proteins by native chemical ligation
WO1997000267A1 (en) 1995-06-16 1997-01-03 Pence Conformationally-restricted combinatorial library composition and method
WO1997013537A1 (en) 1995-10-10 1997-04-17 Visionary Medical Products Corporation Gas pressured needle-less injection device
US5622852A (en) 1994-10-31 1997-04-22 Washington University Bcl-x/Bcl-2 associated cell death regulator
US5650133A (en) 1990-01-19 1997-07-22 Nycomed Salutar Macrocyclic polyaza dichelates linked through ring nitrogens via an amide or ester functionality
WO1997030072A1 (en) 1996-02-20 1997-08-21 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Peptidomimetic inhibitors of cathepsin d and plasmepsins i and ii
US5663316A (en) 1996-06-18 1997-09-02 Clontech Laboratories, Inc. BBC6 gene for regulation of cell death
US5672584A (en) 1995-04-25 1997-09-30 The University Of Kansas Cyclic prodrugs of peptides and peptide nucleic acids having improved metabolic stability and cell membrane permeability
WO1997037705A1 (en) 1996-04-11 1997-10-16 Weston Medical Limited Spring-powered dispensing device for medical purposes
US5708136A (en) 1992-04-07 1998-01-13 The Johns Hopkins University Polypetides which bind to human MDM2
WO1998001467A2 (en) 1996-07-05 1998-01-15 Novartis Ag Inhibitors of the interaction between p53 and mdm2
US5731408A (en) 1995-04-10 1998-03-24 Arizona Board Of Regents On Behalf Of The University Of Arizona Peptides having potent antagonist and agonist bioactivities at melanocortin receptors
US5750767A (en) 1989-10-23 1998-05-12 Research Corporation Technologies, Inc. Synthesis and use of amino acid fluorides as peptide coupling reagents
US5811515A (en) 1995-06-12 1998-09-22 California Institute Of Technology Synthesis of conformationally restricted amino acids, peptides, and peptidomimetics by catalytic ring closing metathesis
US5817752A (en) 1996-06-06 1998-10-06 La Jolla Pharmaceutical Company Cyclic polypeptides comprising a thioether linkage and methods for their preparation
WO1998046631A1 (en) 1997-04-11 1998-10-22 Eli Lilly And Company Combinatorial libraries of peptidomimetic macrocycles and processes therefor
US5840833A (en) 1995-10-27 1998-11-24 Molecumetics, Ltd Alpha-helix mimetics and methods relating thereto
US5851775A (en) 1997-03-20 1998-12-22 Johns Hopkins University β-catenin, Tcf-4, and APC interact to prevent cancer
US5856445A (en) 1996-10-18 1999-01-05 Washington University Serine substituted mutants of BCL-XL /BCL-2 associated cell death regulator
US5874529A (en) 1994-06-08 1999-02-23 Peptor Ltd. Conformationally constrained backbone cyclized peptide analogs
WO1999014259A1 (en) 1997-09-12 1999-03-25 Shearwater Polymers Degradable poly(ethylene glycol) hydrogels with controlled half-life and precursors therefor
US5922863A (en) 1992-04-03 1999-07-13 California Institute Of Technology Diene cyclization using ruthenium and osmium carbene complexes
WO1999034833A1 (en) 1998-01-07 1999-07-15 Shearwater Polymers, Incorporated Degradable heterobifunctional poly(ethylene glycol) acrylates and gels and conjugates derived therefrom
WO1999034850A1 (en) 1998-01-08 1999-07-15 Fiderm S.R.L. Device for controlling the penetration depth of a needle, for application to an injection syringe
US5955593A (en) 1996-09-09 1999-09-21 Washington University BH3 interacting domain death agonist
US5965703A (en) 1996-09-20 1999-10-12 Idun Pharmaceuticals Human bad polypeptides, encoding nucleic acids and methods of use
WO2000006187A2 (en) 1998-07-31 2000-02-10 Washington University Modulation of apoptosis
US6030997A (en) 1998-01-21 2000-02-29 Eilat; Eran Acid labile prodrugs
US6031072A (en) 1996-07-12 2000-02-29 Mcgill University Compounds and methods for modulating cell adhesion
US6043339A (en) 1994-06-13 2000-03-28 Vanderbilt University Method for importing biologically active molecules into cells
US6046289A (en) 1990-11-27 2000-04-04 Komazawa; Hiroyuki Propenamide derivatives containing Arg-Gly-Asp polymers obtained therefrom
US6051554A (en) 1995-06-07 2000-04-18 Peptor Limited Conformationally constrained backbone cyclized somatostatin analogs
US6054556A (en) 1995-04-10 2000-04-25 The Arizona Board Of Regents On Behalf Of The University Of Arizona Melanocortin receptor antagonists and agonists
CN1252808A (en) 1997-02-20 2000-05-10 耶达研究及发展有限公司 Antipathogenic synthetic piptides and compositions comprising them
US6169073B1 (en) 1995-02-16 2001-01-02 Bayer Corporation Peptides and peptidomimetics with structural similarity to human p53 that activate p53 function
US6177542B1 (en) 1993-09-27 2001-01-23 The Burnham Institute Integrin-binding peptides
US6184344B1 (en) 1995-05-04 2001-02-06 The Scripps Research Institute Synthesis of proteins by native chemical ligation
US6204361B1 (en) 1996-01-18 2001-03-20 Research Corporation Technologies, Inc. Method of peptide synthesis
US6271198B1 (en) 1996-11-06 2001-08-07 Genentech, Inc. Constrained helical peptides and methods of making same
US6287787B1 (en) 1993-11-24 2001-09-11 Torrey Pines Institute For Molecular Studies Dimeric oligopeptide mixture sets
US6326354B1 (en) 1998-08-19 2001-12-04 Washington University Modulation of apoptosis with bid
US6348558B1 (en) 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
WO2002064790A2 (en) 2000-12-19 2002-08-22 The Johns Hopkins University Jfy1 protein induces rapid apoptosis
WO2002072597A2 (en) 2001-03-09 2002-09-19 University Of Louisville Helicomimetics and stabilized lxxll peptidomimetics
US6495674B1 (en) 2000-02-25 2002-12-17 The Salk Institute For Biological Studies Evectins and their use
US6569993B1 (en) 1998-04-15 2003-05-27 Aventis Pharma S.A. Process for the preparation of resin-bound cyclic peptides
WO2003059933A2 (en) 2002-01-03 2003-07-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Conformationally constrained c-backbone cyclic peptides
US6610657B1 (en) 1996-11-21 2003-08-26 Promega Corporation Alkyl peptide amides and applications
WO2003070892A2 (en) 2002-02-15 2003-08-28 The Regents Of The University Of Michigan Inhibitors of rgs proteins
US6613874B1 (en) 1999-03-29 2003-09-02 The Procter & Gamble Company Melanocortin receptor ligands
US20030166138A1 (en) 2002-02-21 2003-09-04 Todd Kinsella Cyclic peptides and analogs useful to treat allergies
WO2003106491A2 (en) 2002-06-18 2003-12-24 Cepep Ab Cell penetrating peptides
US6686148B1 (en) 1999-03-01 2004-02-03 Nuvelo, Inc. Methods for targeting RNA molecules
US20040023887A1 (en) 1998-09-02 2004-02-05 Renuka Pillutla Insulin and IGF-1 receptor agonists and antagonists
US20040038901A1 (en) 2000-07-28 2004-02-26 Universitat Zurich Essential downstream component of the wingless signaling pathway and therapeutic and diagnostic applications based thereon
US6703382B2 (en) 2000-08-16 2004-03-09 Georgetown University Medical Center Small molecule inhibitors targeted at Bcl-2
US6713280B1 (en) 1999-04-07 2004-03-30 Thomas Jefferson University Enhancement of peptide cellular uptake
WO2004026896A2 (en) 2002-09-23 2004-04-01 Medivir Ab Hcv ns-3 serine protease inhibitors
US20040067503A1 (en) 2002-04-22 2004-04-08 Weihong Tan Functionalized nanoparticles and methods of use
WO2004041275A1 (en) 2002-11-08 2004-05-21 F. Hoffmann-La Roche Ag Substituted 4-alkoxyoxazol derivatives as ppar agonists
US20040106159A1 (en) 2000-02-22 2004-06-03 Florian Kern Method for antigen-specific stimulation of t-lymphocytes with synthetic peptide libraries
US20040106548A1 (en) 2001-09-07 2004-06-03 Schmidt Michelle A Conformationally constrained labeled peptides for imaging and therapy
US20040115135A1 (en) 2002-12-17 2004-06-17 Quay Steven C. Compositions and methods for enhanced mucosal delivery of peptide YY and methods for treating and preventing obesity
WO2004058804A1 (en) 2002-12-24 2004-07-15 Walter And Eliza Hall Institute Of Medical Research Peptides and therapeutic uses thereof
US20040152708A1 (en) 2002-11-07 2004-08-05 Yong Li Trans-9,10-dehydroepothilone C and D, analogs thereof and methods of making the same
EP1452868A2 (en) 2003-02-27 2004-09-01 Pepscan Systems B.V. Method for selecting a candidate drug compound
US20040171809A1 (en) 2002-09-09 2004-09-02 Korsmeyer Stanley J. BH3 peptides and method of use thereof
US20040235746A1 (en) 1994-06-13 2004-11-25 Hawiger Jack J. Cell permeable peptides for inhibition of inflammatory reactions and methods of use
WO2005007675A2 (en) 2003-07-09 2005-01-27 The Scripps Research Institute TRIAZOLE &epsiv;-AMINO ACIDS
US6849428B1 (en) 1997-03-05 2005-02-01 New England Biolabs, Inc. Intein-mediated protein ligation of expressed proteins
WO2005012335A1 (en) 2003-07-30 2005-02-10 Amersham Health As Imaging agents
CN1583730A (en) 2004-06-04 2005-02-23 中国科学院上海有机化学研究所 Synthesis of 5-iodo-1,4-twice substituted-1,2,3-trioxazole compound
US6875594B2 (en) 1997-11-13 2005-04-05 The Rockefeller University Methods of ligating expressed proteins
WO2005040202A2 (en) 2003-10-16 2005-05-06 Aplagen Gmbh Stabilized alpha-helical peptides
WO2005044839A2 (en) 2003-11-05 2005-05-19 Dana-Farber Cancer Institute, Inc. Stabilized alpha helical peptides and uses thereof
US20050119167A1 (en) 2003-06-02 2005-06-02 Giovanni Abbenante Process for the preparation of cyclic peptides
EP1541692A1 (en) 2002-09-06 2005-06-15 Kaneka Corporation PROCESS FOR PRODUCING L-a-METHYLCYSTEINE DERIVATIVE
WO2005085457A2 (en) 2004-03-03 2005-09-15 University Of Leeds Method and products for the selective degradation of proteins
WO2005090388A1 (en) 2004-03-19 2005-09-29 The University Of Queensland Alpha helical mimics, their uses and methods for their production
US20050222427A1 (en) 2002-05-30 2005-10-06 The Scripps Research Institute Copper-catalysed ligation of azides and acetylenes
EP1602663A1 (en) 2004-06-04 2005-12-07 Chiralix B.V. Triazole-linked glycoamino acids and glycopeptides
WO2005118620A2 (en) 2004-05-27 2005-12-15 New York University Methods for preparing internally constraied peptides and peptidomimetics
WO2005118634A2 (en) 2004-06-04 2005-12-15 The Brigham And Women's Hospital, Inc. Helical peptidomimetics with enhanced activity against beta-amyloid production
US20060008848A1 (en) 1999-05-18 2006-01-12 Verdine Gregory L Stabilized compounds having secondary structure motifs
WO2006038208A2 (en) 2004-07-12 2006-04-13 Medical Research Fund Of Tel Aviv Sourasky Medical Center Agents capable of downregulating an msf-a - dependent hif-1α and use thereof in cancer treatment
US20060111411A1 (en) 2004-10-29 2006-05-25 Schering Corporation Substituted 5-carboxyamide pyrazoles and [1,2,4]triazoles as antiviral agents
US7064193B1 (en) 1997-09-17 2006-06-20 The Walter And Eliza Hall Institute Of Medical Research Therapeutic molecules
US20060148715A1 (en) 2004-12-20 2006-07-06 Baylor College Of Medicine Structural requirements for STAT3 binding and recruitment to phosphotyrosine ligands
WO2006078161A1 (en) 2005-01-24 2006-07-27 Pepscan Systems B.V. Binding compounds, immunogenic compounds and peptidomimetics
US7083983B2 (en) 1996-07-05 2006-08-01 Cancer Research Campaign Technology Limited Inhibitors of the interaction between P53 and MDM2
US7084244B2 (en) 1994-06-08 2006-08-01 Develogen Israel Ltd. Conformationally constrained backbone cyclized peptide analogs
WO2006103666A2 (en) 2005-03-28 2006-10-05 Yeda Research And Development Co. Ltd. Isolated bid polypeptides, polynucleotides encoding same and antibodies directed thereagainst and methods of using same for inducing cell cycle arrest or apoptosis
US20060293380A1 (en) 2003-10-03 2006-12-28 Nantermet Philippe G Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
US20070020620A1 (en) 2005-07-14 2007-01-25 Finn M G Compositions and methods for coupling a plurality of compounds to a scaffold
US7183059B2 (en) 1998-03-23 2007-02-27 President And Fellows Of Harvard College Synthesis of compounds and libraries of compounds
US20070117154A1 (en) 1999-10-04 2007-05-24 Pierre Deslongchamps Combinatorial Synthesis of Libraries of Macrocyclic Compounds Useful in Drug Discovery
US20070161544A1 (en) 2006-01-06 2007-07-12 Peter Wipf Selective targeting agents for mitcochondria
US7247700B2 (en) 2001-12-31 2007-07-24 Dana Farber Cancer Institute, Inc. BID polypeptides and methods of inducing apoptosis
US20070203057A1 (en) 1998-07-10 2007-08-30 Adherex Technologies, Inc. Compounds and methods for modulating adhesion molecule function
WO2007141533A2 (en) 2006-06-09 2007-12-13 Almac Discovery Limited Fkbp-l and uses thereof
WO2007144886A2 (en) 2006-06-15 2007-12-21 Ben-Gurion University Of The Negev Research And Development Authority Virus-like particles for treatment of viral infections
WO2008013454A2 (en) 2006-07-26 2008-01-31 Pepscan Systems B.V. Immunogenic compounds and protein mimics
US20080081831A1 (en) 2000-01-24 2008-04-03 Adherex Technologies, Inc. Peptidomimetic modulators of cell adhesion
WO2008045238A2 (en) 2006-10-05 2008-04-17 New York Blood Center, Inc. Stabilized therapeutic small helical antiviral peptides
WO2008061192A2 (en) 2006-11-15 2008-05-22 Dana-Farber Cancer Institute, Inc. Stabilized maml peptides and uses thereof
WO2008074895A1 (en) 2006-12-21 2008-06-26 Cytos Biotechnology Ag Circular ccr5 peptide conjugates and uses thereof
WO2008076904A1 (en) 2006-12-14 2008-06-26 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
WO2008095063A1 (en) 2007-01-31 2008-08-07 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
WO2008104000A2 (en) 2007-02-23 2008-08-28 Aileron Therapeutics, Inc. Triazole macrocycle systems
US20080213175A1 (en) 2006-09-15 2008-09-04 Kolb Hartmuth C Click chemistry-derived cyclic peptidomimetics as integrin markers
WO2008121767A2 (en) 2007-03-28 2008-10-09 President And Fellows Of Harvard College Stitched polypeptides
WO2008137633A2 (en) 2007-05-02 2008-11-13 Dana-Farber Cancer Institute, Inc. Methods of modulating cellular homeostatic pathways and cellular survival
US20090088553A1 (en) 2006-12-14 2009-04-02 Aileron Therapeutics, Inc., A Delaware Corporation Bis-Sulfhydryl Macrocyclization Systems
WO2009042237A2 (en) 2007-09-26 2009-04-02 Dana Farber Cancer Institute Methods and compositions for modulating bcl-2 family polypeptides
US7538190B2 (en) 2006-02-17 2009-05-26 Polychip Pharmaceuticals Pty Ltd Methods for the synthesis of two or more dicarba bridges in organic compounds
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
WO2009099677A2 (en) 2008-02-08 2009-08-13 Aileron Therapeutics, Inc. Therapeutic peptidomimetic macrocycles
WO2009108261A2 (en) 2008-01-23 2009-09-03 Dana Farber Cancer Institute Compositions and methods for the treatment of viral infections
WO2009110952A2 (en) 2007-12-31 2009-09-11 New York University Control of viral-host membrane fusion with hydrogen bond surrogate-based artificial helices
WO2009126292A2 (en) 2008-04-08 2009-10-15 Aileron Therapeutics, Inc. Biologically active peptidomimetic macrocycles
WO2009149214A2 (en) 2008-06-03 2009-12-10 Aileron Therapeutics, Inc. Compositions and methods for enhancing cellular transport of biomolecules
WO2010011313A2 (en) 2008-07-23 2010-01-28 President And Fellows Of Harvard College Ligation of stapled polypeptides
WO2010034026A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010034031A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010034028A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic marcrocycles
WO2010034034A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010034032A2 (en) 2008-09-22 2010-03-25 Aileron Therapeutic, Inc. Methods for preparing purified polypeptide compositions
WO2010034029A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010033879A2 (en) 2008-09-18 2010-03-25 New York University Inhibiting interaction between hif-1a and p300/cbp with hydrogen bond surrogate-based helices
WO2010060112A1 (en) 2008-11-24 2010-05-27 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with improved properties
WO2010068684A2 (en) 2008-12-09 2010-06-17 Dana Farber Cancer Institute, Inc. Methods and compositions for specific modulation of mcl-1
US7745573B2 (en) 2006-02-17 2010-06-29 Polychip Pharmaceuticals Pty Ltd. Conotoxin analogues and methods for synthesis of intramolecular dicarba bridge-containing peptides
WO2010083347A2 (en) 2009-01-14 2010-07-22 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US7838711B2 (en) 2004-07-08 2010-11-23 Boehringer Ingelheim International Gmbh Process for continuous ruthenium-catalysed metathesis
WO2011008260A2 (en) 2009-07-13 2011-01-20 President And Fellows Of Harvard College Bifunctional stapled polypeptides and uses thereof
US7875601B2 (en) 1998-02-25 2011-01-25 Novartis Ag Cancer treatment with epothilones
US20110065915A1 (en) 2008-01-25 2011-03-17 Trustees Of Boston Coilege Catalysts for metathesis reactions including enantioselective olefin metathesis, and related methods
WO2011038049A1 (en) 2009-09-22 2011-03-31 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2011047215A1 (en) 2009-10-14 2011-04-21 Aileron Therapeutics, Inc. Improved peptidomimetic macrocycles
US7932397B2 (en) 2006-11-22 2011-04-26 Massachusetts Institute Of Technology Olefin metathesis catalysts and related methods
US20110144303A1 (en) 2008-04-08 2011-06-16 Aileron Therapeutics, Inc. Biologically Active Peptidomimetic Macrocycles
US20110245477A1 (en) 2010-02-08 2011-10-06 Trustees Of Boston College Efficient methods for z- or cis-selective cross-metathesis
WO2012021874A1 (en) 2010-08-13 2012-02-16 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with thioether linkers
WO2012040459A2 (en) 2010-09-22 2012-03-29 President And Fellows Of Harvard College Beta-catenin targeting peptides and uses thereof
WO2012122059A1 (en) 2011-03-04 2012-09-13 New York University Hydrogen bond surrogate macrocycles as modulators of ras
WO2012174423A1 (en) 2011-06-17 2012-12-20 President And Fellows Of Harvard College Stabilized polypeptides as regulators of rab gtpase function
WO2012173846A2 (en) 2011-06-06 2012-12-20 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2013033645A1 (en) 2011-08-31 2013-03-07 New York University Thioether-,ether-, and alkylamine-linked hydrogen bond surrogate pertidomimentics
WO2013059525A1 (en) 2011-10-18 2013-04-25 Aileron Therapeutics, Inc. Peptidomimetic macrocyles
WO2013059530A2 (en) 2011-10-18 2013-04-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20130177979A1 (en) 2010-06-22 2013-07-11 University Of Central Florida Research Foundation, Inc. Methods and compositions for cell permeable stat3 inhibitor
WO2013123267A1 (en) 2012-02-15 2013-08-22 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
WO2013123266A1 (en) 2012-02-15 2013-08-22 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2014052647A2 (en) 2012-09-26 2014-04-03 President And Fellows Of Harvard College Proline-locked stapled peptides and uses thereof
WO2014055564A1 (en) 2012-10-01 2014-04-10 President And Fellows Of Harvard College Stabilized polypeptide insulin receptor modulators
US20140128581A1 (en) 2012-11-01 2014-05-08 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US20140256912A1 (en) 2011-06-17 2014-09-11 President And Fellows Of Harvard College Stabilized Variant MAML Peptides and Uses Thereof
WO2014138429A2 (en) 2013-03-06 2014-09-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and use thereof in regulating hif1alpha
WO2015157508A1 (en) 2014-04-09 2015-10-15 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with pth activity
WO2016049359A1 (en) 2014-09-24 2016-03-31 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
WO2016049355A1 (en) 2014-09-24 2016-03-31 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
WO2016154058A1 (en) 2015-03-20 2016-09-29 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
WO2017004548A1 (en) 2015-07-01 2017-01-05 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2017004591A2 (en) 2015-07-02 2017-01-05 Dana-Farber Cancer Institute, Inc. Stabilized anti-microbial peptides
WO2017023933A2 (en) 2015-08-03 2017-02-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2017040990A1 (en) 2015-09-03 2017-03-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
WO2017044633A1 (en) 2015-09-10 2017-03-16 Aileron Therapeutics, Inc. Peptidomimetic macrocycles as modulators of mcl-1
US20170360881A1 (en) 2016-06-17 2017-12-21 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3704108A (en) 1970-09-25 1972-11-28 Hydrocarbon Research Inc Hydroconversion of waste natural and synthetic rubbers

Patent Citations (339)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4000259A (en) 1975-06-16 1976-12-28 American Home Products Corporation Cyclic dodecapeptide analogs of somatostatin and intermediates
US4438270A (en) 1977-07-11 1984-03-20 Merrell Toraude Et Compagnie α-Halomethyl derivatives of α-amino acids
US4191754A (en) 1979-02-28 1980-03-04 Merck & Co., Inc. Bicyclic somatostatin analogs
US4730006A (en) 1986-01-27 1988-03-08 Merrell Dow Pharmaceuticals Inc. Derivatives of 2,6-diamino-3-haloheptanedioic acid
WO1989009233A1 (en) 1988-03-24 1989-10-05 Terrapin Technologies, Inc. Molecular sticks for controlling protein conformation
US5120859A (en) 1989-09-22 1992-06-09 Genentech, Inc. Chimeric amino acid analogues
US5750767A (en) 1989-10-23 1998-05-12 Research Corporation Technologies, Inc. Synthesis and use of amino acid fluorides as peptide coupling reagents
US5650133A (en) 1990-01-19 1997-07-22 Nycomed Salutar Macrocyclic polyaza dichelates linked through ring nitrogens via an amide or ester functionality
US5245009A (en) 1990-03-23 1993-09-14 The Salk Institute For Biological Studies CRF antagonists
EP0467699A2 (en) 1990-07-19 1992-01-22 Merck & Co. Inc. Cyclic HIV principal neutralizing determinant peptides
US6046289A (en) 1990-11-27 2000-04-04 Komazawa; Hiroyuki Propenamide derivatives containing Arg-Gly-Asp polymers obtained therefrom
WO1992013878A2 (en) 1991-02-07 1992-08-20 Board Of Trustees Of The University Of Illinois Conformationally restricted mimetics of beta turns and beta bulges and peptides containing the same
US5364851A (en) 1991-06-14 1994-11-15 International Synthecon, Llc Conformationally restricted biologically active peptides, methods for their production and uses thereof
WO1993001203A1 (en) 1991-07-11 1993-01-21 Smithkline Beecham Plc Antifungal amphotericin derivative
WO1993019176A1 (en) 1992-03-23 1993-09-30 Imperial Cancer Research Technology Limited Leucine zippers
US5922863A (en) 1992-04-03 1999-07-13 California Institute Of Technology Diene cyclization using ruthenium and osmium carbene complexes
US5708136A (en) 1992-04-07 1998-01-13 The Johns Hopkins University Polypetides which bind to human MDM2
WO1994025482A1 (en) 1993-04-23 1994-11-10 Evans Herbert J Polypeptides that include conformation-constraining groups which flank a protein-protein interaction site
US5446128A (en) 1993-06-18 1995-08-29 The Board Of Trustees Of The University Of Illinois Alpha-helix mimetics and methods relating thereto
US5710245A (en) 1993-06-18 1998-01-20 Molecumetics, Ltd. Alpha-helix mimetics and methods relating thereto
WO1995000534A1 (en) 1993-06-18 1995-01-05 The Board Of Trustees Of The University Of Illinois Alpha-helix mimetics and methods relating thereto
US5834209A (en) 1993-08-26 1998-11-10 Washington University Bcl-x/bcl-2 associated cell death regulator
US6177542B1 (en) 1993-09-27 2001-01-23 The Burnham Institute Integrin-binding peptides
US6287787B1 (en) 1993-11-24 2001-09-11 Torrey Pines Institute For Molecular Studies Dimeric oligopeptide mixture sets
US5824483A (en) 1994-05-18 1998-10-20 Pence Inc. Conformationally-restricted combinatiorial library composition and method
US5874529A (en) 1994-06-08 1999-02-23 Peptor Ltd. Conformationally constrained backbone cyclized peptide analogs
US7084244B2 (en) 1994-06-08 2006-08-01 Develogen Israel Ltd. Conformationally constrained backbone cyclized peptide analogs
US20040235746A1 (en) 1994-06-13 2004-11-25 Hawiger Jack J. Cell permeable peptides for inhibition of inflammatory reactions and methods of use
US6043339A (en) 1994-06-13 2000-03-28 Vanderbilt University Method for importing biologically active molecules into cells
WO1996002642A1 (en) 1994-07-20 1996-02-01 University Of Dundee INTERRUPTION OF BINDING OF MDM2 AND p53 PROTEIN AND THERAPEUTIC APPLICATION THEREOF
US6153391A (en) 1994-07-20 2000-11-28 University Of Dundee Interruption of binding of MDM2 and P53 protein and therapeutic application thereof
US5622852A (en) 1994-10-31 1997-04-22 Washington University Bcl-x/Bcl-2 associated cell death regulator
US6169073B1 (en) 1995-02-16 2001-01-02 Bayer Corporation Peptides and peptidomimetics with structural similarity to human p53 that activate p53 function
EP0729972A1 (en) 1995-02-28 1996-09-04 F. Hoffmann-La Roche Ag Peptide derivatives of tetrahydronaphthalene
WO1996028449A1 (en) 1995-03-14 1996-09-19 Hoffman/Barrett, L.L.C. Heteroatom-functionalized porphyrazines and multimetallic complexes and polymers derived therefrom
US5731408A (en) 1995-04-10 1998-03-24 Arizona Board Of Regents On Behalf Of The University Of Arizona Peptides having potent antagonist and agonist bioactivities at melanocortin receptors
US6054556A (en) 1995-04-10 2000-04-25 The Arizona Board Of Regents On Behalf Of The University Of Arizona Melanocortin receptor antagonists and agonists
US5672584A (en) 1995-04-25 1997-09-30 The University Of Kansas Cyclic prodrugs of peptides and peptide nucleic acids having improved metabolic stability and cell membrane permeability
US6184344B1 (en) 1995-05-04 2001-02-06 The Scripps Research Institute Synthesis of proteins by native chemical ligation
WO1996034878A1 (en) 1995-05-04 1996-11-07 The Scripps Research Institute Synthesis of proteins by native chemical ligation
US6051554A (en) 1995-06-07 2000-04-18 Peptor Limited Conformationally constrained backbone cyclized somatostatin analogs
US5811515A (en) 1995-06-12 1998-09-22 California Institute Of Technology Synthesis of conformationally restricted amino acids, peptides, and peptidomimetics by catalytic ring closing metathesis
WO1997000267A1 (en) 1995-06-16 1997-01-03 Pence Conformationally-restricted combinatorial library composition and method
WO1997013537A1 (en) 1995-10-10 1997-04-17 Visionary Medical Products Corporation Gas pressured needle-less injection device
US5840833A (en) 1995-10-27 1998-11-24 Molecumetics, Ltd Alpha-helix mimetics and methods relating thereto
US5859184A (en) 1995-10-27 1999-01-12 Molecumetics Ltd. Alpha-helix mimetics and methods relating thereto
US6204361B1 (en) 1996-01-18 2001-03-20 Research Corporation Technologies, Inc. Method of peptide synthesis
WO1997030072A1 (en) 1996-02-20 1997-08-21 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Peptidomimetic inhibitors of cathepsin d and plasmepsins i and ii
WO1997037705A1 (en) 1996-04-11 1997-10-16 Weston Medical Limited Spring-powered dispensing device for medical purposes
US6031073A (en) 1996-06-06 2000-02-29 La Jolla Pharmaceutical Company Cyclic polypeptides comprising a thioether linkage and methods for their preparation
US5817752A (en) 1996-06-06 1998-10-06 La Jolla Pharmaceutical Company Cyclic polypeptides comprising a thioether linkage and methods for their preparation
US5663316A (en) 1996-06-18 1997-09-02 Clontech Laboratories, Inc. BBC6 gene for regulation of cell death
US7083983B2 (en) 1996-07-05 2006-08-01 Cancer Research Campaign Technology Limited Inhibitors of the interaction between P53 and MDM2
WO1998001467A2 (en) 1996-07-05 1998-01-15 Novartis Ag Inhibitors of the interaction between p53 and mdm2
US6031072A (en) 1996-07-12 2000-02-29 Mcgill University Compounds and methods for modulating cell adhesion
US5998583A (en) 1996-09-09 1999-12-07 Washington University BH3 interacting domain death agonist
US5955593A (en) 1996-09-09 1999-09-21 Washington University BH3 interacting domain death agonist
US5965703A (en) 1996-09-20 1999-10-12 Idun Pharmaceuticals Human bad polypeptides, encoding nucleic acids and methods of use
US5856445A (en) 1996-10-18 1999-01-05 Washington University Serine substituted mutants of BCL-XL /BCL-2 associated cell death regulator
US6271198B1 (en) 1996-11-06 2001-08-07 Genentech, Inc. Constrained helical peptides and methods of making same
US6610657B1 (en) 1996-11-21 2003-08-26 Promega Corporation Alkyl peptide amides and applications
CN1252808A (en) 1997-02-20 2000-05-10 耶达研究及发展有限公司 Antipathogenic synthetic piptides and compositions comprising them
US6849428B1 (en) 1997-03-05 2005-02-01 New England Biolabs, Inc. Intein-mediated protein ligation of expressed proteins
US5851775A (en) 1997-03-20 1998-12-22 Johns Hopkins University β-catenin, Tcf-4, and APC interact to prevent cancer
WO1998046631A1 (en) 1997-04-11 1998-10-22 Eli Lilly And Company Combinatorial libraries of peptidomimetic macrocycles and processes therefor
WO1999014259A1 (en) 1997-09-12 1999-03-25 Shearwater Polymers Degradable poly(ethylene glycol) hydrogels with controlled half-life and precursors therefor
US7064193B1 (en) 1997-09-17 2006-06-20 The Walter And Eliza Hall Institute Of Medical Research Therapeutic molecules
US6875594B2 (en) 1997-11-13 2005-04-05 The Rockefeller University Methods of ligating expressed proteins
WO1999034833A1 (en) 1998-01-07 1999-07-15 Shearwater Polymers, Incorporated Degradable heterobifunctional poly(ethylene glycol) acrylates and gels and conjugates derived therefrom
WO1999034850A1 (en) 1998-01-08 1999-07-15 Fiderm S.R.L. Device for controlling the penetration depth of a needle, for application to an injection syringe
US6030997A (en) 1998-01-21 2000-02-29 Eilat; Eran Acid labile prodrugs
US7875601B2 (en) 1998-02-25 2011-01-25 Novartis Ag Cancer treatment with epothilones
US7183059B2 (en) 1998-03-23 2007-02-27 President And Fellows Of Harvard College Synthesis of compounds and libraries of compounds
US6569993B1 (en) 1998-04-15 2003-05-27 Aventis Pharma S.A. Process for the preparation of resin-bound cyclic peptides
US20070203057A1 (en) 1998-07-10 2007-08-30 Adherex Technologies, Inc. Compounds and methods for modulating adhesion molecule function
WO2000006187A2 (en) 1998-07-31 2000-02-10 Washington University Modulation of apoptosis
JP2002524391A (en) 1998-07-31 2002-08-06 ワシントン ユニヴァーシティー Regulation of apoptosis
US6326354B1 (en) 1998-08-19 2001-12-04 Washington University Modulation of apoptosis with bid
US20040023887A1 (en) 1998-09-02 2004-02-05 Renuka Pillutla Insulin and IGF-1 receptor agonists and antagonists
US20150056612A1 (en) 1999-03-01 2015-02-26 Ling X. Shen Methods for targeting rna molecules
US6686148B1 (en) 1999-03-01 2004-02-03 Nuvelo, Inc. Methods for targeting RNA molecules
US8389484B2 (en) 1999-03-01 2013-03-05 Ling X. Shen Methods for targeting RNA molecules
US7115372B2 (en) 1999-03-01 2006-10-03 Variagenics, Inc. Methods for targeting RNA molecules
US6613874B1 (en) 1999-03-29 2003-09-02 The Procter & Gamble Company Melanocortin receptor ligands
US6713280B1 (en) 1999-04-07 2004-03-30 Thomas Jefferson University Enhancement of peptide cellular uptake
US20110028753A1 (en) 1999-05-18 2011-02-03 President And Fellows Of Harvard College Stabilized Compounds Having Secondary Structure Motifs
US7786072B2 (en) 1999-05-18 2010-08-31 President And Fellows Of Harvard College Stabilized compounds having secondary structure motifs
US20130211046A1 (en) 1999-05-18 2013-08-15 President And Fellows Of Harvard College Stabilized Compounds Having Secondary Structure Motifs
US20060008848A1 (en) 1999-05-18 2006-01-12 Verdine Gregory L Stabilized compounds having secondary structure motifs
US20140162339A1 (en) 1999-05-18 2014-06-12 President And Fellows Of Harvard College Stabilized Compounds Having Secondary Structure Motifs
US20170088581A1 (en) 1999-05-18 2017-03-30 President And Fellows Of Harvard College Stabilized compounds having secondary structure motifs
US8895699B2 (en) 1999-05-18 2014-11-25 President And Fellows Of Harvard College Stabilized compounds having secondary structure motifs
US9505801B2 (en) 1999-05-18 2016-11-29 President And Fellows Of Harvard College Stabilized compounds having secondary structure motifs
US8324428B2 (en) 1999-05-18 2012-12-04 President And Fellows Of Harvard College Stabilized compounds having secondary structure motifs
US7192713B1 (en) 1999-05-18 2007-03-20 President And Fellows Of Harvard College Stabilized compounds having secondary structure motifs
US20070117154A1 (en) 1999-10-04 2007-05-24 Pierre Deslongchamps Combinatorial Synthesis of Libraries of Macrocyclic Compounds Useful in Drug Discovery
US6348558B1 (en) 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
US20080081831A1 (en) 2000-01-24 2008-04-03 Adherex Technologies, Inc. Peptidomimetic modulators of cell adhesion
US20040106159A1 (en) 2000-02-22 2004-06-03 Florian Kern Method for antigen-specific stimulation of t-lymphocytes with synthetic peptide libraries
US6495674B1 (en) 2000-02-25 2002-12-17 The Salk Institute For Biological Studies Evectins and their use
US20040038901A1 (en) 2000-07-28 2004-02-26 Universitat Zurich Essential downstream component of the wingless signaling pathway and therapeutic and diagnostic applications based thereon
US6703382B2 (en) 2000-08-16 2004-03-09 Georgetown University Medical Center Small molecule inhibitors targeted at Bcl-2
WO2002064790A2 (en) 2000-12-19 2002-08-22 The Johns Hopkins University Jfy1 protein induces rapid apoptosis
WO2002072597A2 (en) 2001-03-09 2002-09-19 University Of Louisville Helicomimetics and stabilized lxxll peptidomimetics
US20040106548A1 (en) 2001-09-07 2004-06-03 Schmidt Michelle A Conformationally constrained labeled peptides for imaging and therapy
US7247700B2 (en) 2001-12-31 2007-07-24 Dana Farber Cancer Institute, Inc. BID polypeptides and methods of inducing apoptosis
WO2003059933A2 (en) 2002-01-03 2003-07-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Conformationally constrained c-backbone cyclic peptides
WO2003070892A2 (en) 2002-02-15 2003-08-28 The Regents Of The University Of Michigan Inhibitors of rgs proteins
US20030166138A1 (en) 2002-02-21 2003-09-04 Todd Kinsella Cyclic peptides and analogs useful to treat allergies
US20040067503A1 (en) 2002-04-22 2004-04-08 Weihong Tan Functionalized nanoparticles and methods of use
US20050222427A1 (en) 2002-05-30 2005-10-06 The Scripps Research Institute Copper-catalysed ligation of azides and acetylenes
WO2003106491A2 (en) 2002-06-18 2003-12-24 Cepep Ab Cell penetrating peptides
US20080234183A1 (en) 2002-06-18 2008-09-25 Mattias Hallbrink Cell Penetrating Peptides
EP1541692A1 (en) 2002-09-06 2005-06-15 Kaneka Corporation PROCESS FOR PRODUCING L-a-METHYLCYSTEINE DERIVATIVE
US20040171809A1 (en) 2002-09-09 2004-09-02 Korsmeyer Stanley J. BH3 peptides and method of use thereof
WO2004026896A2 (en) 2002-09-23 2004-04-01 Medivir Ab Hcv ns-3 serine protease inhibitors
US20040152708A1 (en) 2002-11-07 2004-08-05 Yong Li Trans-9,10-dehydroepothilone C and D, analogs thereof and methods of making the same
WO2004041275A1 (en) 2002-11-08 2004-05-21 F. Hoffmann-La Roche Ag Substituted 4-alkoxyoxazol derivatives as ppar agonists
US20040115135A1 (en) 2002-12-17 2004-06-17 Quay Steven C. Compositions and methods for enhanced mucosal delivery of peptide YY and methods for treating and preventing obesity
WO2004058804A1 (en) 2002-12-24 2004-07-15 Walter And Eliza Hall Institute Of Medical Research Peptides and therapeutic uses thereof
US20060073518A1 (en) 2003-02-27 2006-04-06 Pepscan Systems B. V. Method for selecting a candidate drug compound
EP1452868A2 (en) 2003-02-27 2004-09-01 Pepscan Systems B.V. Method for selecting a candidate drug compound
WO2004077062A2 (en) 2003-02-27 2004-09-10 Pepscan Systems B.V. Method for selecting a candidate drug compound
EP1597585B1 (en) 2003-02-27 2011-06-15 Pepscan Systems B.V. Method for selecting a candidate drug compound
US20050119167A1 (en) 2003-06-02 2005-06-02 Giovanni Abbenante Process for the preparation of cyclic peptides
WO2005007675A2 (en) 2003-07-09 2005-01-27 The Scripps Research Institute TRIAZOLE &epsiv;-AMINO ACIDS
WO2005012335A1 (en) 2003-07-30 2005-02-10 Amersham Health As Imaging agents
US20060293380A1 (en) 2003-10-03 2006-12-28 Nantermet Philippe G Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005040202A2 (en) 2003-10-16 2005-05-06 Aplagen Gmbh Stabilized alpha-helical peptides
US8796418B2 (en) 2003-11-05 2014-08-05 Dana-Farber Cancer Institute, Inc. Stabilized alpha helical peptides and uses thereof
US9464115B2 (en) 2003-11-05 2016-10-11 Dana-Farber Cancer Institute, Inc. Stabilized alpha helical peptides and uses thereof
US7723469B2 (en) 2003-11-05 2010-05-25 Dana-Farber Cancer Institute, Inc. Stabilized alpha helical peptides and uses thereof
US20170008930A1 (en) 2003-11-05 2017-01-12 Loren D. Walensky Stabilized alpha helical peptides and uses thereof
WO2005044839A2 (en) 2003-11-05 2005-05-19 Dana-Farber Cancer Institute, Inc. Stabilized alpha helical peptides and uses thereof
US20120082636A1 (en) 2003-11-05 2012-04-05 Walensky Loren D Stabilized alpha helical peptides and uses thereof
US8198405B2 (en) 2003-11-05 2012-06-12 Dana-Farber Cancer Institute, Inc. Stabilized alpha helical peptides and uses thereof
US20050250680A1 (en) 2003-11-05 2005-11-10 Walensky Loren D Stabilized alpha helical peptides and uses thereof
US20090176964A1 (en) 2003-11-05 2009-07-09 Walensky Loren D Stabilized Alpha Helical Peptides and Uses Thereof
US20090149630A1 (en) 2003-11-05 2009-06-11 Walensky Loren D Stabilized Alpha Helical Peptides and Uses Thereof
US9273099B2 (en) 2003-11-05 2016-03-01 President And Fellows Of Harvard College Stabilized alpha helical peptides and uses thereof
US20140296160A1 (en) 2003-11-05 2014-10-02 President And Fellows Of Harvard College Stabilized alpha helical peptides and uses thereof
WO2005085457A2 (en) 2004-03-03 2005-09-15 University Of Leeds Method and products for the selective degradation of proteins
WO2005090388A1 (en) 2004-03-19 2005-09-29 The University Of Queensland Alpha helical mimics, their uses and methods for their production
US8071541B2 (en) 2004-05-27 2011-12-06 Aileron Therapeutics, Inc. Methods for preparing internally constrained peptides and peptidomimetics
US20070197772A1 (en) 2004-05-27 2007-08-23 New York University Methods for preparing internally constrained peptides and peptidomimetics
US20140051828A1 (en) 2004-05-27 2014-02-20 New York University Methods for preparing internally constrained peptides and peptidomimetics
US20060014675A1 (en) 2004-05-27 2006-01-19 Paramjit Arora Methods for preparing internally constrained peptides and peptidomimetics
US20100234563A1 (en) 2004-05-27 2010-09-16 Paramjit Arora Methods for Preparing Internally Constrained Peptides and Peptidomimetics
WO2005118620A2 (en) 2004-05-27 2005-12-15 New York University Methods for preparing internally constraied peptides and peptidomimetics
US7202332B2 (en) 2004-05-27 2007-04-10 New York University Methods for preparing internally constrained peptides and peptidomimetics
US7705118B2 (en) 2004-05-27 2010-04-27 New York University Methods for preparing internally constrained peptides and peptidomimetics
US20130005943A1 (en) 2004-05-27 2013-01-03 Paramjit Arora Methods for Preparing Internally Constrained Peptides and Peptidomimetics
CN1583730A (en) 2004-06-04 2005-02-23 中国科学院上海有机化学研究所 Synthesis of 5-iodo-1,4-twice substituted-1,2,3-trioxazole compound
EP1602663A1 (en) 2004-06-04 2005-12-07 Chiralix B.V. Triazole-linked glycoamino acids and glycopeptides
WO2005118625A1 (en) 2004-06-04 2005-12-15 Chiralix B.V. Triazole-linked glycoamino acids and glycopeptides
WO2005118634A2 (en) 2004-06-04 2005-12-15 The Brigham And Women's Hospital, Inc. Helical peptidomimetics with enhanced activity against beta-amyloid production
US7838711B2 (en) 2004-07-08 2010-11-23 Boehringer Ingelheim International Gmbh Process for continuous ruthenium-catalysed metathesis
WO2006038208A2 (en) 2004-07-12 2006-04-13 Medical Research Fund Of Tel Aviv Sourasky Medical Center Agents capable of downregulating an msf-a - dependent hif-1α and use thereof in cancer treatment
US20060111411A1 (en) 2004-10-29 2006-05-25 Schering Corporation Substituted 5-carboxyamide pyrazoles and [1,2,4]triazoles as antiviral agents
US20060148715A1 (en) 2004-12-20 2006-07-06 Baylor College Of Medicine Structural requirements for STAT3 binding and recruitment to phosphotyrosine ligands
WO2006078161A1 (en) 2005-01-24 2006-07-27 Pepscan Systems B.V. Binding compounds, immunogenic compounds and peptidomimetics
WO2006103666A2 (en) 2005-03-28 2006-10-05 Yeda Research And Development Co. Ltd. Isolated bid polypeptides, polynucleotides encoding same and antibodies directed thereagainst and methods of using same for inducing cell cycle arrest or apoptosis
US20070020620A1 (en) 2005-07-14 2007-01-25 Finn M G Compositions and methods for coupling a plurality of compounds to a scaffold
US20070161544A1 (en) 2006-01-06 2007-07-12 Peter Wipf Selective targeting agents for mitcochondria
US7745573B2 (en) 2006-02-17 2010-06-29 Polychip Pharmaceuticals Pty Ltd. Conotoxin analogues and methods for synthesis of intramolecular dicarba bridge-containing peptides
US8124726B2 (en) 2006-02-17 2012-02-28 Monash University Contoxin analogues and methods for synthesizing same
US7538190B2 (en) 2006-02-17 2009-05-26 Polychip Pharmaceuticals Pty Ltd Methods for the synthesis of two or more dicarba bridges in organic compounds
WO2007141533A2 (en) 2006-06-09 2007-12-13 Almac Discovery Limited Fkbp-l and uses thereof
WO2007144886A2 (en) 2006-06-15 2007-12-21 Ben-Gurion University Of The Negev Research And Development Authority Virus-like particles for treatment of viral infections
US20100291040A1 (en) 2006-06-15 2010-11-18 Ben-Gu-Rion University Of The Negev Research And Development Authority Virus-like particles for treatment of viral infections
WO2008013454A2 (en) 2006-07-26 2008-01-31 Pepscan Systems B.V. Immunogenic compounds and protein mimics
US20080213175A1 (en) 2006-09-15 2008-09-04 Kolb Hartmuth C Click chemistry-derived cyclic peptidomimetics as integrin markers
WO2008045238A2 (en) 2006-10-05 2008-04-17 New York Blood Center, Inc. Stabilized therapeutic small helical antiviral peptides
US20100081611A1 (en) 2006-11-15 2010-04-01 James Bradner Stabilized maml peptides and uses thereof
WO2008061192A2 (en) 2006-11-15 2008-05-22 Dana-Farber Cancer Institute, Inc. Stabilized maml peptides and uses thereof
US7932397B2 (en) 2006-11-22 2011-04-26 Massachusetts Institute Of Technology Olefin metathesis catalysts and related methods
US9675661B2 (en) 2006-12-14 2017-06-13 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US8609809B2 (en) 2006-12-14 2013-12-17 Aileron Thraputics, Inc. Bis-sulfhydryl macrocyclization systems
US20160095896A1 (en) 2006-12-14 2016-04-07 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US20170296620A1 (en) 2006-12-14 2017-10-19 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US20100184628A1 (en) 2006-12-14 2010-07-22 Aileron Therapeutics, Inc., A Delaware Corporation Bis-sulfhydryl macrocyclization systems
US20090088553A1 (en) 2006-12-14 2009-04-02 Aileron Therapeutics, Inc., A Delaware Corporation Bis-Sulfhydryl Macrocyclization Systems
US20090047711A1 (en) 2006-12-14 2009-02-19 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US7960506B2 (en) 2006-12-14 2011-06-14 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
WO2008076904A1 (en) 2006-12-14 2008-06-26 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US9175056B2 (en) 2006-12-14 2015-11-03 Alleron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US7981998B2 (en) 2006-12-14 2011-07-19 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
WO2008074895A1 (en) 2006-12-21 2008-06-26 Cytos Biotechnology Ag Circular ccr5 peptide conjugates and uses thereof
WO2008095063A1 (en) 2007-01-31 2008-08-07 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
US20150119551A1 (en) 2007-01-31 2015-04-30 President And Fellows Of Harvard College Stabilized p53 peptides and uses thereof
US20170081379A1 (en) 2007-01-31 2017-03-23 Dana-Farber Cancer Institute, Inc. Stabilized P53 Peptides and Uses Thereof
US9527896B2 (en) 2007-01-31 2016-12-27 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
US20100168388A1 (en) 2007-01-31 2010-07-01 Federico Bernal Stabilized p53 peptides and uses thereof
US8889632B2 (en) 2007-01-31 2014-11-18 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
US20160031936A1 (en) 2007-02-23 2016-02-04 Aileron Therapeutics, Inc. Triazole macrocycle systems
US9493509B2 (en) 2007-02-23 2016-11-15 Aileron Therapeutics, Inc. Triazole macrocycle systems
US9023988B2 (en) 2007-02-23 2015-05-05 Aileron Therapeutics, Inc. Triazole macrocycle systems
US20160289274A1 (en) 2007-02-23 2016-10-06 Aileron Therapeutics, Inc. Triazole macrocycle systems
US20160304564A1 (en) 2007-02-23 2016-10-20 Aileron Therapeutics, Inc. Triazole macrocycle systems
WO2008104000A2 (en) 2007-02-23 2008-08-28 Aileron Therapeutics, Inc. Triazole macrocycle systems
US20110263815A1 (en) 2007-02-23 2011-10-27 Aileron Therapeutics, Inc., A Delaware Corporation Triazole macrocycle systems
US7981999B2 (en) 2007-02-23 2011-07-19 Aileron Therapeutics, Inc. Triazole macrocycle systems
US8637686B2 (en) 2007-02-23 2014-01-28 Aileron Therapeutics, Inc. Triazole macrocycle systems
US20080262200A1 (en) 2007-02-23 2008-10-23 Aileron Therapeutics, Inc., A Delaware Corporation Triazole Macrocycle Systems
US20100184645A1 (en) 2007-03-28 2010-07-22 President And Fellows Of Harvard College Stitched polypeptides
US8592377B2 (en) 2007-03-28 2013-11-26 President And Fellows Of Harvard College Stitched polypeptides
US20140011979A1 (en) 2007-03-28 2014-01-09 President And Fellows Of Harvard College Stitched polypeptides
US20170066799A1 (en) 2007-03-28 2017-03-09 President And Fellows Of Harvard College Stitched polypeptides
US9556227B2 (en) 2007-03-28 2017-01-31 President And Fellows Of Harvard College Stitched polypeptides
WO2008121767A2 (en) 2007-03-28 2008-10-09 President And Fellows Of Harvard College Stitched polypeptides
US20100273704A1 (en) 2007-05-02 2010-10-28 Dana-Farber Cancer Institute, Inc. Methods of modulating cellular homeostatic pathways and cellular survival
WO2008137633A2 (en) 2007-05-02 2008-11-13 Dana-Farber Cancer Institute, Inc. Methods of modulating cellular homeostatic pathways and cellular survival
WO2009042237A2 (en) 2007-09-26 2009-04-02 Dana Farber Cancer Institute Methods and compositions for modulating bcl-2 family polypeptides
US8871899B2 (en) 2007-12-31 2014-10-28 New York University Control of viral-host membrane fusion with hydrogen bond surrogate-based artificial helices
US20110046043A1 (en) 2007-12-31 2011-02-24 Deyun Wang Control of viral-host membrane fusion with hydrogen bond surrogate-based artificial helices
WO2009110952A2 (en) 2007-12-31 2009-09-11 New York University Control of viral-host membrane fusion with hydrogen bond surrogate-based artificial helices
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
WO2009108261A2 (en) 2008-01-23 2009-09-03 Dana Farber Cancer Institute Compositions and methods for the treatment of viral infections
US20110065915A1 (en) 2008-01-25 2011-03-17 Trustees Of Boston Coilege Catalysts for metathesis reactions including enantioselective olefin metathesis, and related methods
US8808694B2 (en) 2008-02-08 2014-08-19 Aileron Therapeutics, Inc. Therapeutic peptidomimetic macrocycles
US20090275519A1 (en) 2008-02-08 2009-11-05 Aileron Therapeutics, Inc. Therapeutic peptidomimetic macrocycles
WO2009099677A2 (en) 2008-02-08 2009-08-13 Aileron Therapeutics, Inc. Therapeutic peptidomimetic macrocycles
US20150038430A1 (en) 2008-02-08 2015-02-05 Aileron Therapeutics, Inc. Therapeutic peptidomimetic macrocycles
US20090326192A1 (en) 2008-04-08 2009-12-31 Aileron Therapeutics, Inc. Biologically active peptidomimetic macrocycles
US20160108089A1 (en) 2008-04-08 2016-04-21 Aileron Therapeutics, Inc. Biologically active peptidomimetic macrocycles
US20140323701A1 (en) 2008-04-08 2014-10-30 Aileron Therapeutics, Inc. Biologically active peptidomimetic macrocycles
WO2009126292A2 (en) 2008-04-08 2009-10-15 Aileron Therapeutics, Inc. Biologically active peptidomimetic macrocycles
US20110144303A1 (en) 2008-04-08 2011-06-16 Aileron Therapeutics, Inc. Biologically Active Peptidomimetic Macrocycles
US20130023646A1 (en) 2008-04-08 2013-01-24 Aileron Therapeutics, Inc. Biologically active peptidomimetic macrocycles
US20170298099A1 (en) 2008-04-08 2017-10-19 Aileron Therapeutics, Inc. Biologically active peptidomimetic macrocycles
WO2009149214A2 (en) 2008-06-03 2009-12-10 Aileron Therapeutics, Inc. Compositions and methods for enhancing cellular transport of biomolecules
US20110250685A1 (en) 2008-06-03 2011-10-13 Nash Huw M Compositions and methods for enhancing cellular transport of biomolecules
US20110144306A1 (en) 2008-07-23 2011-06-16 President And Fellows Of Harvard College Ligation of stapled polypeptides
WO2010011313A2 (en) 2008-07-23 2010-01-28 President And Fellows Of Harvard College Ligation of stapled polypeptides
WO2010033879A2 (en) 2008-09-18 2010-03-25 New York University Inhibiting interaction between hif-1a and p300/cbp with hydrogen bond surrogate-based helices
US20110245175A1 (en) 2008-09-18 2011-10-06 New York University INHIBITING INTERACTION BETWEEN THE HIF-1ALPHA AND p300/CBP WITH HYDROGEN BOND SURROGATE-BASED
WO2010034026A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9175045B2 (en) 2008-09-22 2015-11-03 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20120264674A1 (en) 2008-09-22 2012-10-18 Aileron Therapeutics, Inc. Methods for preparing purified polypeptide compositions
US20120178700A1 (en) 2008-09-22 2012-07-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20120172311A1 (en) 2008-09-22 2012-07-05 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8524653B2 (en) 2008-09-22 2013-09-03 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20120115783A1 (en) 2008-09-22 2012-05-10 Alleron therapeutics, Inc Peptidomimetic macrocycles
US20120115793A1 (en) 2008-09-22 2012-05-10 Alleron therapeutics, Inc Peptidomimetic macrocycles
US20120101047A1 (en) 2008-09-22 2012-04-26 Aileron Therapetics Inc. Peptidomimetic macrocycles
US20160096873A1 (en) 2008-09-22 2016-04-07 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8399405B2 (en) 2008-09-22 2013-03-19 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9394336B2 (en) 2008-09-22 2016-07-19 Aileron Therapeutics, Inc. Methods for preparing purified polypeptide compositions
US9206223B2 (en) 2008-09-22 2015-12-08 Aileron Therapeutics, Inc. Methods for preparing purified polypeptide compositions
US20100210515A1 (en) 2008-09-22 2010-08-19 Aileron Therapeutics, Inc. Methods for preparing purified polypeptide compositions
WO2010034031A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010034028A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic marcrocycles
US20100216688A1 (en) 2008-09-22 2010-08-26 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010034034A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010034032A2 (en) 2008-09-22 2010-03-25 Aileron Therapeutic, Inc. Methods for preparing purified polypeptide compositions
US20160115204A1 (en) 2008-09-22 2016-04-28 Aileron Therapeutics, Inc. Methods for preparing purified polypeptide compositions
WO2010034029A1 (en) 2008-09-22 2010-03-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20100298201A1 (en) 2008-11-24 2010-11-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with improved properties
WO2010060112A1 (en) 2008-11-24 2010-05-27 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with improved properties
US20170212125A1 (en) 2008-11-24 2017-07-27 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with improved properties
US9458202B2 (en) 2008-11-24 2016-10-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with improved properties
WO2010068684A2 (en) 2008-12-09 2010-06-17 Dana Farber Cancer Institute, Inc. Methods and compositions for specific modulation of mcl-1
US20160068573A1 (en) 2009-01-14 2016-03-10 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20120040889A1 (en) 2009-01-14 2012-02-16 Aileron Therapeutics Inc. Peptidomimetic macrocycles
US9175047B2 (en) 2009-01-14 2015-11-03 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20160251399A1 (en) 2009-01-14 2016-09-01 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2010083347A2 (en) 2009-01-14 2010-07-22 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2011008260A2 (en) 2009-07-13 2011-01-20 President And Fellows Of Harvard College Bifunctional stapled polypeptides and uses thereof
US9163330B2 (en) 2009-07-13 2015-10-20 President And Fellows Of Harvard College Bifunctional stapled polypeptides and uses thereof
US20120270800A1 (en) 2009-07-13 2012-10-25 President And Fellows Of Harvard College Bifunctional stapled polypeptides and uses thereof
US20130072439A1 (en) 2009-09-22 2013-03-21 Huw M. Nash Peptidomimetic macrocycles
WO2011038049A1 (en) 2009-09-22 2011-03-31 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20170266254A1 (en) 2009-09-22 2017-09-21 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20110223149A1 (en) 2009-10-14 2011-09-15 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2011047215A1 (en) 2009-10-14 2011-04-21 Aileron Therapeutics, Inc. Improved peptidomimetic macrocycles
US20110245477A1 (en) 2010-02-08 2011-10-06 Trustees Of Boston College Efficient methods for z- or cis-selective cross-metathesis
US20130177979A1 (en) 2010-06-22 2013-07-11 University Of Central Florida Research Foundation, Inc. Methods and compositions for cell permeable stat3 inhibitor
WO2012021876A2 (en) 2010-08-13 2012-02-16 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2012021874A1 (en) 2010-08-13 2012-02-16 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with thioether linkers
US20140378390A1 (en) 2010-08-13 2014-12-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8859723B2 (en) 2010-08-13 2014-10-14 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2012021875A1 (en) 2010-08-13 2012-02-16 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with triazole linkers
US20160257716A1 (en) 2010-08-13 2016-09-08 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20150284437A1 (en) 2010-09-22 2015-10-08 President And Fellows Of Harvard College Beta-catenin targeting peptides and uses thereof
US20140005118A1 (en) 2010-09-22 2014-01-02 President And Fellows Of Harvard College Beta-catenin targeting peptides and uses thereof
US8957026B2 (en) 2010-09-22 2015-02-17 President And Fellows Of Harvard College Beta-catenin targeting peptides and uses thereof
WO2012040459A2 (en) 2010-09-22 2012-03-29 President And Fellows Of Harvard College Beta-catenin targeting peptides and uses thereof
WO2012122059A1 (en) 2011-03-04 2012-09-13 New York University Hydrogen bond surrogate macrocycles as modulators of ras
WO2012173846A2 (en) 2011-06-06 2012-12-20 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20140235549A1 (en) 2011-06-17 2014-08-21 President And Fellows Of Harvard College Stabilized polypeptides as regulators of rab gtpase function
WO2012174423A1 (en) 2011-06-17 2012-12-20 President And Fellows Of Harvard College Stabilized polypeptides as regulators of rab gtpase function
US20140256912A1 (en) 2011-06-17 2014-09-11 President And Fellows Of Harvard College Stabilized Variant MAML Peptides and Uses Thereof
US20130123196A1 (en) 2011-08-31 2013-05-16 New York University Thioether-, ether-, and alkylamine-linked hydrogen bond surrogate peptidomimetics
WO2013033645A1 (en) 2011-08-31 2013-03-07 New York University Thioether-,ether-, and alkylamine-linked hydrogen bond surrogate pertidomimentics
US20170226177A1 (en) 2011-10-18 2017-08-10 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9522947B2 (en) 2011-10-18 2016-12-20 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2013059530A2 (en) 2011-10-18 2013-04-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2013059525A1 (en) 2011-10-18 2013-04-25 Aileron Therapeutics, Inc. Peptidomimetic macrocyles
US20160137710A1 (en) 2011-10-18 2016-05-19 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8927500B2 (en) 2012-02-15 2015-01-06 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2013123266A1 (en) 2012-02-15 2013-08-22 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20150183825A1 (en) 2012-02-15 2015-07-02 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
WO2013123267A1 (en) 2012-02-15 2013-08-22 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US8987414B2 (en) 2012-02-15 2015-03-24 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US20170281720A1 (en) 2012-02-15 2017-10-05 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20150051155A1 (en) 2012-02-15 2015-02-19 Aileron Therapeutics Peptidomimetic macrocycles
US9505804B2 (en) 2012-02-15 2016-11-29 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2014052647A2 (en) 2012-09-26 2014-04-03 President And Fellows Of Harvard College Proline-locked stapled peptides and uses thereof
US20150239937A1 (en) 2012-09-26 2015-08-27 President And Fellows Of Harvard College Proline-locked stapled peptides and uses thereof
US20150225471A1 (en) 2012-10-01 2015-08-13 President And Fellows Of Harvard College Stabilized polypeptide insulin receptor modulators
WO2014055564A1 (en) 2012-10-01 2014-04-10 President And Fellows Of Harvard College Stabilized polypeptide insulin receptor modulators
US20170066714A1 (en) 2012-11-01 2017-03-09 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US9845287B2 (en) 2012-11-01 2017-12-19 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
WO2014071241A1 (en) 2012-11-01 2014-05-08 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US20140128581A1 (en) 2012-11-01 2014-05-08 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US9604919B2 (en) 2012-11-01 2017-03-28 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
WO2014138429A2 (en) 2013-03-06 2014-09-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and use thereof in regulating hif1alpha
US20160052970A1 (en) 2013-03-06 2016-02-25 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and use thereof in regulating hif1alpha
WO2015157508A1 (en) 2014-04-09 2015-10-15 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with pth activity
US20170037086A1 (en) 2014-04-09 2017-02-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles with pth activity
US20160101145A1 (en) 2014-09-24 2016-04-14 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
WO2016049359A1 (en) 2014-09-24 2016-03-31 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
WO2016049355A1 (en) 2014-09-24 2016-03-31 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
US20160193283A1 (en) 2014-09-24 2016-07-07 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US20160333049A1 (en) 2015-03-20 2016-11-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
WO2016154058A1 (en) 2015-03-20 2016-09-29 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
WO2017004548A1 (en) 2015-07-01 2017-01-05 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20170002042A1 (en) 2015-07-01 2017-01-05 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
WO2017004591A2 (en) 2015-07-02 2017-01-05 Dana-Farber Cancer Institute, Inc. Stabilized anti-microbial peptides
US20170015716A1 (en) 2015-07-02 2017-01-19 Dana-Farber Cancer Institute, Inc. Stabilized anti-microbial peptides
WO2017023933A2 (en) 2015-08-03 2017-02-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20170037105A1 (en) 2015-08-03 2017-02-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US20170114098A1 (en) 2015-09-03 2017-04-27 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
WO2017040990A1 (en) 2015-09-03 2017-03-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US20170107252A1 (en) 2015-09-10 2017-04-20 Aileron Therapeutics, Inc. Peptidomimetic macrocycles as modulators of mcl-1
WO2017044633A1 (en) 2015-09-10 2017-03-16 Aileron Therapeutics, Inc. Peptidomimetic macrocycles as modulators of mcl-1
US20170360881A1 (en) 2016-06-17 2017-12-21 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
WO2017218949A2 (en) 2016-06-17 2017-12-21 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof

Non-Patent Citations (493)

* Cited by examiner, † Cited by third party
Title
"Fustero, et al. Asymmetric synthesis of new beta,beta-difluorinated cyclic quaternary alpha-amino acid derivatives. Org Lett. Aug. 31, 2006;8(18):4129-32."
"Sidwell, R. W. et al., In vitro and in vivo assay systems for study of influenza virus inhibitors. Antiviral Res, 2000. 48(1): p. 1-16".
Andrews et al. Forming Stable Helical Peptide Using Natural and Artificial Amino Acids. Tetrahedron. 1999;55:11711-11743.
Angel & Karin, "The Role of Jun, Fos and the AP-1 Complex in Cell-proliferation and Transformation," Biochim. Biophys. Acta 1072:129-157 (1991).
Angell, et al. Peptidomimetics via copper-catalyzed azide-alkyne cycloadditions. Chem Soc Rev. Oct. 2007;36(10):1674-89.
Angell, et al. Ring closure to beta-turn mimics via copper-catalyzed azide/alkyne cycloadditions. J Org Chem. Nov. 11, 2005;70(23):9595-8.
Annis, et al. A general technique to rank protein-ligand binding affinities and determine allosteric versus direct binding site competition in compound mixtures. J Am Chem Soc. Dec. 1, 2004;126(47):15495-503.
Annis, et al. ALIS: An affinity selection-mass spectrometry system for the discovery and characterization of protein-ligand Interactions. Mass Spectrometry in Medicinal Chemistry: Applications in Drug Discovery (2007): 121-156.
Armstrong et al., X = Y-ZH Systems as potential 1,3-dipoles. 5. Intramolecular cycloadditions of imines of a-amino acid esters. Tetrahedron. 1985;41(17):3547-58.
Arora, "Design, Synthesis, and Properties of the Hydrogen Bond Surrogate-based Artificial Alpha-helices," American Chemical Society Meeting, San Diego (Mar. 2005) (oral).
Arora, "Hydrogen Bond Surrogate Approach for the Synthesis of Short α-Helical Peptides," American Chemical Society Meeting, Philadelphia (Aug. 2004) (abstract of oral presentation).
Arosio, et al. Click chemistry to functionalise peptidomimetics. Tetrahedron Letters. 2006; 47:3697-3700.
Austin et al., "A Template for Stabilization of a Peptide α-Helix: Synthesis and Evaluation of Conformational Effects by Circular Dichroism and NMR," J. Am. Chem. Soc. 119:6461-6472 (1997).
Babine et aL, Molecular Recognition of Proteinminus signLigand Complexes: Applications to Drug Design. Chem Rev. Aug. 5, 1997;97(5):1359-1472.
Baell, J.B. Prospects for Targeting the Bcl-2 Family of Proteins to Develop Novel cytotoxic drugs. Biochem Pharmacol. Sep. 2002;64(5-6):851-63.
Bagnasco, et al. Inhibition of a protein-protein interaction between INI1 and c-Myc by small peptidomimetic molecules inspired by Helix-1 of c-Myc: identification of a new target of potential antineoplastic interest. FASEB J. Apr. 2007;21(4):1256-63. Epub Jan. 10, 2007.
Bakhshi, et al. Cloning the chromosomal breakpoint of t(14;18) human lymphomas: clustering around JH on chromosome 14 and near a transcriptional unit on 18. Cell. Jul. 1985;41(3):899-906.
Banerji et al. Synthesis of Cyclic β-Turn Mimics from L-Pro-Phe/Phe-L-Pro Derived Di- and Tripeptides via Ring Closing Metathesis: The Role of Chirality of the Phe Residue During Cyclization. Tetrahedron Lett. 2002; 43:6473-6477.
Bang et al., Total chemical synthesis of crambin. J Am Chem Soc. Feb. 11, 2004;126(5):1377-83.
Barker, et al. Cyclic RGD peptide analogues as antiplatelet antithrombotics. J Med Chem. May 29, 1992;35(11):2040-8. (Abstract only).
Belokon et al., Chiral Complexes of Ni(II), Cu(II) and Cu(I) as Reagents, Catalysts and Receptors for Asymmetric Synthesis and Chiral Recognition of Amino Acids. Pure & Appl Chem. 1992;64(12):1917-24.
Belokon, et al. Improved procedures for the synthesis of (S)-2-[N-(N′- benzylprolyl)amino]benzophenone (BPB) and Ni(II) complexes of Schiff's bases derived from BPB and amino acids. Tetrahedron: Asymmetry, vol. 9, Issue 23, Dec. 11, 1998, pp. 4249-4252.
Berendsen et al. A glimpse of the Holy Grail? Science 282(5389):642-643 (1998).
Berge et al. Pharmaceutical Salts. Journal of Pharmaceutical Sciences 66(1):1-19 (Jan. 1977).
Bernal, et al. Reactivation of the p53 tumor suppressor pathway by a stapled p53 peptide. J Am Chem Soc. Mar. 7, 2007;129(9):2456-7.
Biagini et al., Cross-metathesis of Unsaturated a-amino Acid Derivatives. J Chem Soc Perkin Trans. 1998;1:2485-99.
Bierzynski et al. A salt bridge stabilizes the helix formed by isolated C-Peptide of RNase A. PNAS USA. 1982;79:2470-2474.
Blackwell, et al. Highly Efficient Synthesis of Covalently Cross-Linked Peptide Helices by Ring-Closing Metatheis. Angewandte Chemie International Edition. 1998; 37(23):3281-3284.
Blackwell, et al. Ring-closing metathesis of olefinic peptides: design, synthesis, and structural characterization of macrocyclic helical peptides. J Org Chem. Aug. 10, 2001;66(16):5291-302.
Boal et al.,Facile and E-Selective Intramolecular Ring-Closing Metathesis Reactions in 310-Helical Peptides: A 3D Structural Study. J. Am. Chem. Soc. 2007, 129, 6986-6987.
Boal, et al. Facile and E-selective intramolecular ring-closing metathesis reactions in 3(10)-helical peptides: a 3D structural study. J Am Chem Soc. Jun. 6, 2007;129(22):6986-7. Epub May 11, 2007.
Bock, et al. 1,2,3-Triazoles as peptide bond isosteres: synthesis and biological evaluation of cyclotetrapeptide mimics. Org Biomol Chem. Mar. 21, 2007;5(6):971-5.
Boguslavsky, et al. Effect of peptide conformation on membrane permeability. J Pept Res. Jun. 2003;61(6):287-97.
Bossy-Wetzel et al. Assays for cytochrome c release from mitochondria during apoptosis. Methods Enzymol. 322:235-242 (2000).
Bossy-Wetzel, et al. Detection of apoptosis by annexin V labeling. Methods Enzymol. 2000;322:15-8.
Bracken et al. Synthesis and nuclear magnetic resonance structure determination of an alpha-helical, bicyclic, lactam-bridged hexapeptide. JACS. 1994;116:6431-6432.
Bradley et al. Limits of cooperativity in a structurally modular protein: response of the Notch ankyrin domain to analogous alanine substitutions in each repeat. J Mol Biol. 324(2):373-386 (2002).
Brea, et al. Synthesis of omega-(hetero)arylalkynylated alpha-amino acid by Sonogashira-type reactions in aqueous media. J Org Chem. Sep. 29, 2006;71(20):7870-3.
Brunel, et al. Synthesis of constrained helical peptides by thioether ligation: application to analogs of gp41. Chem Commun (Camb). May 28, 2005;(20):2552-4. Epub Mar. 11, 2005.
Brunel, et al. Synthesis of constrained helical peptides by thioether ligation: application to analogs of gp41. Chemical communications. 2005;20:2552-2554.
Burfield & Smithers, "Desiccant Efficiency in Solvent Drying. 3. Dipolar Aprotic Solvents," J. Org. Chem. 43(20):3966-3968 (1978).
Burrage, et al. Biomimetic synthesis of lantibiotics. Chemistry. Apr. 14, 2000;6(8):1455-66.
Cabezas & Satterthwait, "The Hydrogen Bond Mimic Approach: Solid-phase Synthesis of a Peptide Stabilized as an α-Helix with a Hydrazone Link," J. Am. Chem. Soc. 121:3862-3875 (1999).
Campbell, et al. N-alkylated oligoamide alpha-helical proteomimetics. Org Biomol Chem. May 21, 2010;8(10):2344-51. doi: 10.1039/c001164a. Epub Mar. 18, 2010.
Cantel, et al. Synthesis and Conformational Analysis of a Cyclic Peptide Obtained via i to i+4 Intramolecular Side-Chain to Side-Chain Azide-Alkyne 1,3-Dipolar Cycloaddition. JOC Featured Article. Published on the web May 20, 2008.
Carlson et al., Specificity landscapes of DNA binding molecules elucidate biological function. Proc Natl Acad Sci USA. Mar. 9, 2010;107(10):4544-9. doi: 10.1073/pnas.0914023107. Epub Feb. 22, 2010.
CAS Registry No. 2176-37-6, STN Entry Date Nov. 16, 1984.
CAS Registry No. 2408-85-7, STN Entry Date Nov. 16, 1984.
CAS Registry No. 4727-05-3, STN Entry Date Nov. 16, 1984.
CAS Registry No. 561321-72-0, STN Entry Date Aug. 6, 2003.
CAS Registry No. 721918-14-5, STN Entry Date Aug. 4, 2004.
Chakrabartty et al., "Helix Capping Propensities in Peptides Parallel Those in Proteins," Proc. Nat'l Acad. Sci. USA 90:11332-11336 (1993).
Chakrabartty et al., "Helix Propensities of the Amino Acids Measured in Alanine-based Peptides without Helix-stabilizing Side-chain Interactions," Protein Sci. 3:843-852 (1994).
Chang et al. Stapled α-helical peptide drug development: A potent dual inhibitor of MDM2 and MDMX for p53-dependent cancer therapy PNAS 2013 110 (36) E3445-E3454; published ahead of print Aug. 14, 2013, doi:10.1073/pnas.1303002110.
Chapman et al., "A Highly Stable Short α-Helix Constrained by a Main-chain Hydrogen-bond Surrogate," J. Am. Chem. Soc. 126:12252-12253 (2004).
Chapman, et al. Optimized synthesis of hydrogen-bond surrogate helices: surprising effects of microwave heating on the activity of Grubbs catalysts. Org Lett. Dec. 7, 2006;8(25):5825-8.
Chen et al., "Structure of the DNA-binding Domains from NFAT, Fos and Jun Bound Specifically to DNA," Nature 392:42-48 (1998).
Chen et al., Determination of the helix and beta form of proteins in aqueous solution by circular dichroism. Biochemistry. Jul. 30, 1974;13(16):3350-9.
Chen, et al. Determination of the Secondary Structures of Proteins by Circular Dichroism and Optical Rotatory Dispersion. Biochemistry. 1972; 11(22):4120-4131.
Chène et al., "Study of the Cytotoxic Effect of a Peptidic Inhibitor of the p53-hdm2 Interaction in Tumor Cells," FEBS Lett. 529:293-297 (2002).
Chène, P., "Inhibiting the p53-MDM2 Interaction: An Important Target for Cancer Therapy," Nat Rev. Cancer 3:102-109 (2003).
Chin & Schepartz, "Design and Evolution of a Miniature Bcl-2 Binding Protein," Angew. Chem. Int. Ed. 40(20):3806-3809 (2001).
Chin et al., "Circular Dichroism Spectra of Short, Fixed-nucleus Alanine Helices," Proc. Nat'l Acad. Sci. USA 99(24):15416-15421 (2002).
Chittenden, et al. A conserved domain in Bak, distinct from BH1 and BH2, mediates cell death and protein binding functions. EMBO J. Nov. 15, 1995;14(22):5589-96.
Choi, et al. Application of azide-alkyne cycloaddition ‘click chemistry’ for the synthesis of Grb2 SH2 domain-binding macrocycles. Bioorg Med Chem Lett. Oct. 15, 2006;16(20):5265-9.
Choi, et al. Application of azide-alkyne cycloaddition 'click chemistry' for the synthesis of Grb2 SH2 domain-binding macrocycles. Bioorg Med Chem Lett. Oct. 15, 2006;16(20):5265-9.
Chu, et al. Peptide-formation on cysteine-containing peptide scaffolds. Orig Life Evol Biosph. Oct. 1999;29(5):441-9.
Clark et al., Supramolecular Design by Covalent Capture. Design of a Peptide Cylinder via Hydrogen-Bond-Promoted Intermolecular Olefin Metathesis. J Am Chem Soc. 1995;117:12364-65.
Cleary, et al. Nucleotide sequence of a t(14;18) chromosomal breakpoint in follicular lymphoma and demonstration of a breakpoint-cluster region near a transcriptionally active locus on chromosome 18. Proc Natl Acad Sci U S A. Nov. 1985;82(21):7439-43.
Cline, et al. Effects of As(III) binding on alpha-helical structure. J Am Chem Soc. Mar. 12, 2003;125(10):2923-9.
Colacino, et al. Evaluation of the anti-influenza virus activities of 1,3,4-thiadiazol-2-ylcyanamide (LY217896) and its sodium salt. Antimicrob Agents Chemother. Nov. 1990;34(11):2156-63.
Colaluca et al., NUMB controls p53 tumour suppressor activity. Nature. Jan. 3, 2008;451(7174):76-80. doi: 10.1038/nature06412.
Co-pending U.S. Appl. No. 13/494,846, filed Jun. 12, 2012.
Co-pending U.S. Appl. No. 13/655,442, filed Oct. 18, 2010.
Co-pending U.S. Appl. No. 15/201,235, filed Jul. 1, 2016.
Co-pending U.S. Appl. No. 15/229,517, filed Aug. 5, 2016.
Co-pending U.S. Appl. No. 15/233,796, filed Aug. 10, 2016.
Co-pending U.S. Appl. No. 15/240,505, filed Aug. 18, 2016.
Co-pending U.S. Appl. No. 15/256,130, filed Sep. 2, 2016.
Co-pending U.S. Appl. No. 15/257,807, filed Sep. 6, 2016.
Co-pending U.S. Appl. No. 15/259,947, filed Sep. 8, 2016.
Co-pending U.S. Appl. No. 15/278,824, filed Sep. 28, 2016.
Co-pending U.S. Appl. No. 15/287,513, filed Oct. 6, 2016.
Co-pending U.S. Appl. No. 15/332,492, filed Oct. 24, 2016.
Co-pending U.S. Appl. No. 15/349,478, filed Nov. 11, 2016.
Co-pending U.S. Appl. No. 15/463,826, filed Mar. 20, 2017.
Co-pending U.S. Appl. No. 15/493,301, filed Apr. 21, 2017.
Co-pending U.S. Appl. No. 15/592,517, filed May 11, 2017.
Co-pending U.S. Appl. No. 15/625,672, filed Jun. 16, 2017.
Co-pending U.S. Appl. No. 15/711,576, filed Sep. 21, 2017.
Cory et al., "The Bcl-2 Family: Roles in Cell Survival and Oncogenesis," Oncogene 22:8590-8607 (2003).
Cox et al., Insulin receptor expression by human prostate cancers. Prostate. Jan. 1, 2009;69(1):33-40. doi: 10.1002/pros.20852.
Cusack et al. 2,4,6-Tri-isopropylbenzenesulphonyl Hydrazide: A convenient source of Di-Imide. Tetrahedron. 1976;32:2157-2162.
Dames, et al. Structural basis for Hif-1 alpha /CBP recognition in the cellular hypoxic response. Proc Natl Acad Sci U S A. Apr. 16, 2002;99(8):5271-6.
Danial, et al. Cell death: critical control points. Cell. 2004; 116:204-219.
Darnell, Transcription factors as targets for cancer therapy. Nat Rev Cancer. Oct. 2002;2(10):740-9.
Daugherty & Gellman, "A Fluorescence Assay for Leucine Zipper Dimerization: Avoiding Unintended Consequences of Fluorophore Attachment," J. Am. Chem. Soc. 121:4325-4333 (1999).
Definition of Analog from http://cancerweb.ncl.ac.uk/cgi-bin/omd?query=analog. pp. 1-5. Accessed Jul. 7, 2005.
Degterev et al. Identification of Small-molecule Inhibitors of Interaction between the BH3 Domain and Bcl-xL. Nature Cell Biol. 3:173-182 (2001).
Deiters, et al. Adding amino acids with novel reactivity to the genetic code of Saccharomyces cerevisiae. J Am Chem Soc. Oct. 1, 2003;125(39):11782-3.
Deng, et al. Cross-Coupling Reaction of lodo-1,2,3-triazoles Catalyzed by Palladium. Synthesis 2005(16): 2730-2738.
Denmark et al., Cyclopropanation with Diazomethane and Bis(oxazoline)palladium(II) Complexes. J Org Chem. May 16, 1997;62(10):3375-3389.
Designing Custom Peptide. SIGMA Genosys (pp. 1-2) (Accessed Dec. 16, 2004).
Dimartino et al, "A General Approach for the Stabilization of Peptide Secondary Structures," American Chemical Society Meeting, New York (Sep. 2003) (poster).
Dimartino et al. Solid-phase synthesis of hydrogen-bond surrogate-derived alpha-helices. Org Lett. Jun. 9, 2005 9;7(12):2389-92.
Draeger, et al. Interaction of the bHLH-zip domain of c-Myc with H1-type peptides. Characterization of helicity in the H1 peptides by NMR. J Biol Chem. Jan. 21, 1994;269(3):1785-93.
Duronio, Insulin receptor is phosphorylated in response to treatment of HepG2 cells with insulin-like growth factor I. Biochem J. Aug. 15, 1990;270(1):27-32.
Eckert & Kim, "Mechanisms of Viral Membrane Fusion and Its Inhibition," Annu. Rev. Biochem. 70:777-810 (2001).
Ellis et al., Design, synthesis, and evaluation of a new generation of modular nucleophilic glycine equivalents for the efficient synthesis of sterically constrained alpha-amino acids. J Org Chem. Oct. 27, 2006;71(22):8572-8.
Erez, et al. Induction of apoptosis in cultured endothelial cells by a cadherin antagonist peptide: involvement of fibroblast growth factor receptor-mediated signalling. Exp Cell Res. Apr. 1, 2004;294(2):366-78. Abstract only.
Erlanson, et al. Facile synthesis of cyclic peptides containing di-, tri-, tetra-, and Pentasulfides. Tetrahedron Letters. 1998; 39(38):6799-6802.
European office action dated Aug. 20, 2012 for EP Application No. 09730445.5.
Evans et al., The Rise of Azide—Alkyne 1,3-Dipolar ‘Click’ Cycloaddition and its Application to Polymer Science and Surface Modification. Australian Journal of Chemistry. 2007;60:384-95.
Evans et al., The Rise of Azide-Alkyne 1,3-Dipolar 'Click' Cycloaddition and its Application to Polymer Science and Surface Modification. Australian Journal of Chemistry. 2007;60:384-95.
Extended European Search Report for EP 09800675.2, dated Dec. 6, 2012.
Felix et al., "Synthesis, Biological Activity and Conformational Analysis of Cyclic GRF Analogs," Int. J. Pep. Protein Res. 32:441-454 (1988).
Feng et al. Solid-phase SN2 macrocyclization reactions to form beta-turn mimics. Org Lett. Jul. 15, 1999;1(1):121-4.
Fields, et al. Chapter 3 in Synthetic Peptides: A User's Guide. Grant W.H. Freeman & Co. New York, NY. 1992. p. 77.
Fieser, et al. Fieser and Fieser's Reagents for Organic Synthesis. John Wiley and Sons. 1994.
File Hcaplus on STN. AN No. 1990:532752. Burger et al. Synthesis of a-(trifluoromethyl)-substituted a-amino acids. Part 7. An efficient synthesis for a-trifluoromethyl-substituted w-carboxy a-amino acids. Chemiker-Zeitung (1990), 114(3), 101-4. Abstract only, date Oct. 1990.
Fischer, et al. Apoptosis-based therapies and drug targets. Cell Death and Differentiation. 2005; 12:942-961.
Folkers, et al. Methods and principles in medicinal chemistry. Eds. R. Mannhold, H. Kubinyi, and H. Timmerman. Wiley-VCH, 2001.
Formaggio et al., Inversion of 3(10)-helix screw sense in a (D-alpha Me)Leu homo-tetrapeptide induced by a guest D-(alpha Me)Val residue. J Pept Sci. Nov.-Dec. 1995;1(6):396-402.
Freedman, et al. Structural basis for recruitment of CBP/p300 by hypoxia-inducible factor-1 alpha. Proc Natl Acad Sci U S A. Apr. 16, 2002;99(8):5367-72.
Fulda, et al. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene. Aug. 7, 2006;25(34):4798-811.
Furstner et al., Alkyne Metathesis: Development of a Novel Molybdenum-Based Catalyst System and Its Application to the Total Synthesis of Epothilone A and C. Chem Euro J. 2001 ;7(24):5299-5317.
Furstner, et al. Mo[N(t-Bu)(AR)]3 Complexes as catalyst precursors: In situ activation and application to metathesis reactions of alkynes and diynes. J Am chem Soc. 1999; 121:9453-54.
Furstner, et al. Nozaki-Hiyama-Kishi reactions catalytic in chromium. J Am Chem Soc. 1996; 118:12349-57.
Furstner, et al. Nozaki—Hiyama—Kishi reactions catalytic in chromium. J Am Chem Soc. 1996; 118:12349-57.
Galande, et al. An effective method of on-resin disulfide bond formation in peptides. J Comb Chem. Mar.-Apr. 2005;7(2):174-7.
Galande, et al. An effective method of on-resin disulfide bond formation in peptides. Journal of combinatorial chemistry. 2005;7(2):174-177.
Galande, et al. Thioether side chain cyclization for helical peptide formation: inhibitors of estrogen receptor-coactivator interactions. Journal of Peptide Research. 2004; 63(3): 297-302.
Gallivan et al., A neutral, water-soluble olefin metathesis catalyst based on an N-heterocyclic carbene ligand. Tetrahedron Letters. 2005;46:2577-80.
Galluzzi, et al. Guidelines for the use and interpretation of assays for monitoring cell death in higher eukaryotes. Cell Death Differ. Aug. 2009;16(8):1093-107. Epub Apr. 17, 2009.
Gante, Peptidomimetics-Tailored Enzyme Inhibitors. J Angew Chem Int Ed Engl. 1994;33:1699-1720.
Gante, Peptidomimetics—Tailored Enzyme Inhibitors. J Angew Chem Int Ed Engl. 1994;33:1699-1720.
García-Echeverría et al., "Discovery of Potent Antagonists of the Interaction between Human Double Minute 2 and Tumor Suppressor p53," J. Med. Chem. 43:3205-3208 (2000).
Geistlinger & Guy, "An Inhibitor of the Interaction of Thyroid Hormone Receptor β and Glucocorticoid Interacting Protein 1," J. Am. Chem. Soc. 123:1525-1526 (2001).
Gemperli et al., "Paralog-selective Ligands for Bcl-2 Proteins," J. Am. Chem. Soc. 127:1596-1597 (2005).
Gentle et al., Direct production of proteins with N-terminal cysteine for site-specific conjugation. Bioconjug Chem. May-Jun. 2004;15(3):658-63.
Ghadiri & Choi, "Secondary Structure Nucleation in Peptides. Transition Metal Ion Stabilized α-Helices," J. Am. Chem. Soc. 112:1630-1632 (1990).
Ghanem, et al. Peptide-mediated interference with influenza A virus polymerase. J Virol. Jul. 2007;81(14):7801-4.
Giannis et aL, Peptidomimetics for Receptor Ligands-Discovery, Development, and Medical Perspectives. Angew Chem Int Ed Engl. 1993;32:1244-67.
Giannis et aL, Peptidomimetics for Receptor Ligands—Discovery, Development, and Medical Perspectives. Angew Chem Int Ed Engl. 1993;32:1244-67.
Giorello, et al. Inhibition of cancer cell growth and c-Myc transcriptional activity by a c-Myc helix 1-type peptide fused to an internalization sequence. Cancer Res. Aug. 15, 1998;58(16):3654-9.
Glover & Harrison, "Crystal Structure of the Heterodimeric bZIP Transcription Factor c- Fos-c-Jun Bound to DNA," Nature 373:257-261 (1995).
Goncalves, et al. On-resin cyclization of peptide ligands of the Vascular Endothelial Growth Factor Receptor 1 by copper(I)-catalyzed 1,3-dipolar azide-alkyne cycloaddition. Bioorg Med Chem Lett. Oct. 15, 2007;17(20):5590-4.
Goodson et al., Potential Growth Antagonists. I. Hydantoins and Disubstituted Glycines. J Org Chem. 1960;25:1920-24.
Grandori, et al. The Myc/Max/Mad network and the transcriptional control of cell behavior. Annu Rev Cell Dev Biol. 2000;16:653-99.
Green, T.W.; Wuts, P.G.M. Protective Groups in Organic Synthesis, 2nd ed. New York; John Wiley and Sons, Inc.; 1991.
Greene, et al. Protective Groups in Organic Synthesis, 2nd Ed. John Wiley and Sons. 1991.
Greenfield et al. Computed circular dichroism spectra for the evaluation of protein conformation. Biochemistry. Oct. 8, 1969;(10):4108-4116.
Greenlee et al., A General Synthesis of a-vinyl-a-amino acids. Tetrahedron Letters. 1978;42:3999-40002.
Grubbs, et al. Ring-Closing Metathesis and Related Processes in Organic Synthesis. Acc. Chem. Res., 1995, 28 (11), pp. 446-452.
Guinn et al., Synthesis and characterization of polyamides containing unnatural amino acids. Biopolymers. May 1995;35(5):503-12.
Gupta et al., Long-term effects of tumor necrosis factor-alpha treatment on insulin signaling GUPTA pathway in HepG2 cells and HepG2 cells overexpressing constitutively active Akt/PKB. J Cell Biochem. Feb. 15, 2007;100(3):593-607.
Hanessian, et al. Structure-based design and synthesis of macroheterocyclic peptidomimetic inhibitors of the aspartic protease beta-site amyloid precursor protein cleaving enzyme (BACE). J Med Chem. Jul. 27, 2006;49(15):4544-67.
Hara, S. et al. ‘Synthetic studies on halopeptins, anti-inflammatory cyclodepsipeptides’, Peptide Science. 2006 (vol. date 2005), 42nd, pp. 39-42.
Hara, S. et al. 'Synthetic studies on halopeptins, anti-inflammatory cyclodepsipeptides', Peptide Science. 2006 (vol. date 2005), 42nd, pp. 39-42.
Hase; et al., "1,6-Aminosuberic acid analogs of lysine- and arginine-vasopressin and -vasotocin. Synthesis and biological properties. J Am Chem Soc. May 17, 1972;94(10):3590-600."
He, et al. Crystal structure of the polymerase PA(C)-PB1(N) complex from an avian influenza H5N1 virus. Nature. Aug. 28, 2008;454(7208):1123-6. Epub Jul. 9, 2008.
He, X. et al., Crystal structure of the polymerase PAC-PB1N complex from an avian influenza H5N1 virus. Nature, 2008. 454: p. 1123-6.
He, X. et al., Crystal structure of the polymerase PAC—PB1N complex from an avian influenza H5N1 virus. Nature, 2008. 454: p. 1123-6.
Hecht, S.M., ed. Bioorganic Chemistry: Peptides and Proteins. Oxford University Press. New York; 1998.
Hein, et al. Copper(I)-Catalyzed Cycloaddition of Organic Azides and 1-lodoalkynes. Angew Chem Int Ed Engl. 2009;48(43):8018-21.
Hermerka, et al. Detection and characterization of influenza A virus PA-PB2 interaction through a biomolecular fluorescence complementation assay. J Virol. Apr. 2009;83(8):3944-55. Epub Feb. 4, 2009.
Hewitson, et al. The HIF pathway as a therapeutic target. Drug Discov Today. Aug. 15, 2004;9(16):704-11.
Hiroshige, et al. Palladium-mediated macrocyclisations on solid support and its applica- tions to combinatorial synthesis. J. Am. Chem. Soc. 1995; 117:11590-11591.
Horne, et al. Heterocyclic peptide backbone modifications in an alpha-helical coiled coil. J Am Chem Soc. Dec. 1, 2004;126(47):15366-7.
Horne, et al. Structural and biological mimicry of protein surface recognition by alpha/beta-peptide foldamers. Proc Natl Acad Sci U S A. Sep. 1, 2009;106(35):14751-6. doi: 10.1073/pnas.0902663106. Epub Aug. 17, 2009.
Hoveyda et al., "Ru Complexes Bearing Bidentate Carbenes: From Innocent Curiosity to Uniquely Effective Catalysts for Olefin Metathesis," Org. Biomolec. Chem. 2:8-23 (2004).
Hunt, S. The Non-Protein Amino Acids. In: Barrett G.C., ed. Chemistry and Biochemistry of the Amino Acids. New York; Chapman and Hall; 1985.
International Preliminary Report on Patentability for PCT/US2008/058575 dated Oct. 8, 2009.
International Preliminary Report on Patentability for PCT/US2009/004260 dated Feb. 3, 2011.
International Preliminary Report on Patentability for PCT/US2010/001952 dated Jan. 26, 2012.
International Preliminary Report on Patentability for PCT/US2011/052755, dated Apr. 4 2013.
International Preliminary Report on Patentability for PCT/US2012/042719, dated Jan. 3, 2014.
International Preliminary Report on Patentability for PCT/US2012/042738, dated Jan. 3, 2014.
International Preliminary Report on Patentability for PCT/US2013/062004, dated Apr. 9, 2015.
International Preliminary Report on Patentability for PCT/US2013/062929, dated Apr. 16, 2015.
International Preliminary Report on Patentability for PCT/US2014/025544, dated Sep. 24, 2015.
International search report and written opinion dated Feb. 16, 2010 for PCT Application No. US2009/057927.
International search report and written opinion dated Jan. 7, 2011 for PCT Application No. US2010/049892.
International search report and written opinion dated Jul. 6, 2010 for PCT Application No. US2010/021091.
International search report and written opinion dated Mar. 10, 2010 for PCT Application No. US2009/057930.
International search report and written opinion dated Mar. 5, 2010 for PCT Application No. US2009/057928.
International search report and written opinion dated Mar. 8, 2010 for PCT Application No. US09/057925.
International search report and written opinion dated May 16, 2008 for PCT Application No. US2007/084838.
International Search Report and Written Opinion for PCT/US2008/052580, dated May 16, 2008.
International Search Report and Written Opinion for PCT/US2008/058575 dated Nov. 17, 2008.
International Search Report and Written Opinion for PCT/US2009/004260 dated Oct. 15, 2010.
International Search Report and Written Opinion for PCT/US2010/001952 dated Feb. 2, 2011.
International Search Report and Written Opinion for PCT/US2011/052755 dated Apr. 25, 2012.
International Search Report and Written Opinion for PCT/US2012/042719, dated Nov. 1, 2012.
International Search Report and Written Opinion for PCT/US2012/042738, dated Oct. 18, 2012.
International Search Report and Written Opinion for PCT/US2013/062004, dated Apr. 23, 2014.
International Search Report and Written Opinion for PCT/US2013/062929, dated Jan. 30, 2014.
International Search Report and Written Opinion for PCT/US2014/025544, dated Sep. 10, 2014.
International Search Report and Written Opinion for PCT/US2014/058680, dated Apr. 23, 2015.
International search report dated Apr. 28, 2008 for PCT Application No. US2007/87615.
International search report dated May 11, 2006 for PCT Application No. US2005/016894.
International search report dated May 18, 2005 for PCT Application No. US2004/38403.
International search report dated Nov. 30, 2009 for PCT Application No. US2009/02225.
International Search Report dated Sep. 10, 2014 for PCT Application No. US2014/025544.
International search report dated Sep. 25, 2008 for PCT Application No. US2008/54922.
International Search Report with Opinion dated Dec. 12, 2016 for PCT/US16/40744.
Jackson et al. General approach to the synthesis of short alpha-helical peptides. JACS. 1991;113:9391-9392.
Jimi, et al. Selective inhibition of NF-kappa B blocks osteoclastogenesis and prevents inflammatory bone destruction in vivo. Nat Med. Jun. 2004;10(6):617-24. Epub May 23, 2004.
Jin, et al. Structure-based design, synthesis, and activity of peptide inhibitors of RGS4 GAP activity. Methods Enzymol. 2004;389:266-77.
Jin, et al. Structure-based design, synthesis, and pharmacologic evaluation of peptide RGS4 inhibitors. J Pept Res. Feb. 2004;63(2):141-6.
Johannesson, et al. Vinyl sulfide cyclized analogues of angiotensin II with high affinity and full agonist activity at the AT(1) receptor. J Med Chem. Apr. 25, 2002;45(9):1767-77.
Kanan et al. Reaction discovery enabled by DNA-templated synthesis and in vitro selection. Nature. Sep. 30, 2004;431(7008):545-9.
Karle, et al. Structural charateristics of alpha-helical peptide molecules containing Aib residues. Biochemistry. Jul. 24, 1990;29(29):6747-56.
Karle. Flexibility in peptide molecules and restraints imposed by hydrogen bonds, the Aib residue, and core inserts. Biopolymers. 1996;40(1):157-80.
Karwoski et al., Lysinonorleucine cross-link formation in alpha amino heptenoic acid-substituted peptide derivatives. Biopolymers. 1978;17(5):1119-27.
Kaul & Balaram, "Stereochemical Control of Peptide Folding," Bioorg. Med. Chem. 7:105-117 (1999).
Kazmaier, Sythesis of Quaternary Amino Acids Containing 13, y- as well as 7,6-Unsaturated Side Chains via Chelate-Enolate Claisen Rearrangement. Tetrahedron Letters. 1996;37(30):5351-4.
Kedrowski, B.L. et al. ‘Thiazoline ring formation from 2-methylcysteines and 2-halomethylalanines’, Heterocycles. 2002, vol. 58, pp. 601-634.
Kedrowski, B.L. et al. 'Thiazoline ring formation from 2-methylcysteines and 2-halomethylalanines', Heterocycles. 2002, vol. 58, pp. 601-634.
Kelso et al., "A Cyclic Metallopeptide Induces α Helicity in Short Peptide Fragments of Thermolysin," Angew. Chem. Int. Ed. 42(4):421-424 (2003).
Kelso et al., "α-Turn Mimetics: Short Peptide α-Helices Composed of Cyclic Metallopentapeptide Modules," J. Am. Chem. Soc. 126:4828-4842 (2004).
Kemp et al., "Studies of N-Terminal Templates for α-Helix Formation. Synthesis and Conformational Analysis of (2S,5S,8S,11S)-1-Acetyl-1,4-diaza-3-keto-5-carboxy-10-thiatricyclo[2.8.1.04,8]-tridecane (Ac-Hel1-OH)," J. Org. Chem. 56:6672-6682 (1991).
Kemp et al., "Studies of N-Terminal Templates for α-Helix Formation. Synthesis and Conformational Analysis of Peptide Conjugates of (25,5S,8S,11S)-1-Acetyl-1,4-diaza-3-keto-5-carboxy-10-thiatricyclo[2.8.1.04,8]-tridecane (Ac-Hel1-OH)," J. Org. Chem. 56:6683-6697 (1991).
Kent. Advanced Biology. Oxford University Press. 2000.
Khalil et al., An efficient and high yield method for the N-tert-butoxycarbonyl protection of sterically hindered amino acids. Tetrahedron Lett. 1996;37(20):3441-44.
Kiessling, et al. Selective inhibition of c-Myc/Max dimerization and DNA binding by small molecules. Chem Biol. Jul. 2006;13(7):745-51.
Kilby et al., "Potent Suppression of HIV-1 Replication in Humans by T-20, a Peptide Inhibitor of gp41-Mediated Virus Entry," Nat. Med. 4(11):1302-1307 (1998).
Kim et al., Introduction of all-hydrocarbon i,i+3 staples into alpha-helices via ring-closing olefin metathesis. Org Lett. Jul. 2, 2010;12(13):3046-9. doi: 10.1021/011010449.
Kim et al., Stereochemical effects of all-hydrocarbon tethers in i,i+4 stapled peptides. Bioorg Med Chem Lett. May 1, 2009;19(9):2533-6. Epub Mar. 13, 2009.
Kim et al., Synthesis of all-hydrocarbon stapled a-helical peptides by ring-closing olefin metathesis. Nat Protoc. Jun. 2011;6(6):761-71. doi: 10.1038/nprot.2011.324. Epub May 12, 2011.
Kimmerlin et al., ‘100 years of peptide synthesis’: ligation methods for peptide and protein synthesis with applications to beta-peptide assemblies. J Pept Res. Feb. 2005;65(2):229-60.
Kimmerlin et al., '100 years of peptide synthesis': ligation methods for peptide and protein synthesis with applications to beta-peptide assemblies. J Pept Res. Feb. 2005;65(2):229-60.
Kolb et al., Click Chemistry: Diverse Chemical Function from a Few Good Reactions. Angew Chem Int Ed Engl. Jun. 1, 2001;40(11):2004-2021.
Kotha et al., Modification of constrained peptides by ring-closing metathesis reaction. Bioorg Med Chem Lett. Jun. 4, 2001;11(11):1421-3.
Kouzarides, Acetylation: a regulatory modification to rival phosphorylation? EMBO J. Mar. 15, 2000;19(6):1176-9.
Kritzer et al., "Helical β-Peptide Inhibitors of the p53-hDM2 Interaction," J. Am. Chem. Soc. 126:9468-9469 (2004).
Kudaj, et al. An efficient synthesis of optically pure alpha-alkyl-beta-azido- and alpha-alkyl-beta-aminoalanines via ring opening of 3-amino-3-alkyl-2-oxetanones. Tetrahedron Letters. 2007; 48:6794-6797.
Kussie et al, "Structure of the MDM2 Oncoprotein Bound to the p53 Tumor Suppressor Transactivation Domain," Science 274:948-953 (1996).
Kutzki et al., "Development of a Potent Bcl-xL Antagonist Based on α-Helix Mimicry," J. Am. Chem. Soc. 124:11838-11839 (2002).
Kwon, et al. Quantitative comparison of the relative cell permeability of cyclic and linear peptides. Chem Biol. Jun. 2007;14(6):671-7.
Lacombe et al. Reduction of olefins on solid support using diimide. Tetrahedron Letters. 1998;39:6785-6786.
Larock, R.C. Comprehensive Organic Transformations, New York: VCH Publishers; 1989.
Latini, et al. 395 Blocking the interaction between HIF-1alpha and p300 by a 32 amino acid fragment of p35srj inhibits the hypoxia induced transcriptional activity of HIF-1alpha in human U87MG glioma cells. Euro J Canc Suppl. 2004; 2(8):118.
Leduc et al., Helix-stabilized cyclic peptides as selective inhibitors of steroid receptor-coactivator interactions. Proc Natl Acad Sci USA. 2003;100(20):11273-78.
Lee, et al. A novel BH3 ligand that selectively targets Mcl-1 reveals that apoptosis can proceed without Mcl-1 degradation. J Cell Biol. Jan. 28, 2008;180(2):341-355.
Letai, et al. Distinct BH3 Domains Either Sensitize or Activate Mitochondrial Apoptosis, Serving as Prototype Cancer Therapeutics. Cancer Cell. 2002; 2:183-192.
Li, et al. A convenient preparation of 5-iodo-1,4-disubstituted-1,2,3-triazole: multicomponent one-pot reaction of azide and alkyne mediated by Cul-NBS. J Org Chem. May 2, 2008;73(9):3630-3. doi: 10.1021/jo800035v. Epub Mar. 22, 2008.
Li, et al. Structure-based design of thioether-bridged cyclic phosphopeptides binding to Grb2-SH2 domain. Bioorg Med Chem Lett. Mar. 10, 2003;13(5):895-9.
Lifson & Roig, "On the Theory of Helix-coil Transition in Polypeptides," J. Chem. Phys. 34(6):1963-1974 (1961).
Liskamp, et al. Conformationally restricted amino acids and dipeptides, (non)peptidomimetics and secondary structure mimetics. Recl Travl Chim Pays-Bas. 1994; 113:1-19.
Litowski & Hodges, "Designing Heterodimeric Two-stranded α-Helical Coiled-coils: Effects of Hydrophobicity and α-Helical Propensity on Protein Folding, Stability, and Specificity," J. Biol. Chem. 277(40):37272-37279 (2002).
Liu et al., Chemical Ligation Approach to Form a Peptide Bond between Unprotected Peptide Segments. Concept and Model Study. J Am Chem Soc. 1994;116(10):4149-53.
Luo, et al. Mechanism of helix induction by trifluoroethanol: a framework for extrapolating the helix-forming properties of peptides from trifluoroethanol/water mixtures back to water. Biochemistry. Jul. 8, 1997;36(27):8413-21.
Lyu et al, "α-Helix Stabilization by Natural and Unnatural Amino Acids with Alkyl Side Chains," Proc. Nat'l Acad. Sci. USA 88:5317-5320 (1991).
Lyu, et al. Capping Interactions in Isolated α Helices: Position-dependent Substitution Effects and Structure of a Serine-capped Peptide Helix. Biochemistry. 1993; 32:421-425.
Mai, et al. A proapoptotic peptide for the treatment of solid tumors. Cancer Research. 2001; 61:7709-7712.
Maillard, et al. Mastermind critically regulates Notch-mediated lymphoid cell fate decisions. Blood. Sep. 15, 2004;104(6):1696-702. Epub Jun. 8, 2004.
Mangold, et al. Azidoalanine mutagenicity in Salmonella: effect of homologation and alpha-Mutat Res. Feb. 1989;216(1):27-33.methyl substitution.
Mannhold, R et al. Molecular Drug Properties: Measurement and Prediction (Methods and Principles in Medicinal Chemistry). Wiley-VCH; 2007.
Marqusee & Baldwin, "Helix Stabilization by Glu- . . . Lys+ Salt Bridges in Short Peptides of De Novo Design," Proc. Nat'l Acad. Sci. USA 84:8898-8902 (1987).
Marshall et al., Back to the future: ribonuclease A. Biopolymers. 2008;90(3):259-77.
Martin, et al. Thermal [2+2] intramolecular cycloadditions of fuller-1,6-enynes. Angew Chem Int Ed Engl. Feb. 20, 2006;45(9):1439-42.
May, et al. Selective inhibition of NF-kappaB activation by a peptide that blocks the interaction of NEMO with the IkappaB kinase complex. Science. Sep. 1, 2000;289(5484):1550-4.
McGahon, et al. The end of the (cell) line: methods for the study of apoptosis in vitro. Methods Cell Biol. 1995;46:153-85.
McNamara et al. Peptides constrained by an aliphatic linkage between two C(alpha) sites: design, synthesis, and unexpected conformational properties of an i,(i + 4)-linked peptide. J Org Chem. Jun. 29, 2001;66(13):4585-95.
Miller & Scanlan, "oNBS-SPPS: A New Method for Solid-phase Peptide Synthesis," J. Am. Chem. Soc. 120:2690-2691 (1998).
Miller et al., Application of Ring-Closing Metathesis to the Synthesis of Rigidified Amino Acids and Peptides. J Am Chem Soc. 1996;118(40):9606-9614.
Miller et al., Synthesis of Conformationally Restricted Amino Acids and Peptides Employing Olefin Metathesis. J Am Chem Soc. 1995;117(21):5855-5856.
Moellering et al., Abstract 69. Computational modeling and molecular optimization of stabilized alpha-helical peptides targeting NOTCH-CSL transcriptional complexes. Nov. 2010; 8(7):30. DOI: 10.1016/S1359-6349(10)71774-2. Abstract Only, European Journal of Cancer Supplements, 2010, 8(7).
Moellering et al., Direct inhibition of the NOTCH transcription factor complex. Nature. Nov. 12, 2009;462(7270):182-8. Erratum in: Nature. Jan. 21, 2010;463(7279):384.
Mosberg, et al. Dithioeter-containing cyclic peptides. J. Am. Chem. Soc. 1985;107(10):2986-2987.
Mosberg, et al. Dithioether-containing cyclic peptides. Journal of the American Chemical Society. 1985;107(10):2986-2987.
Muchmore, et al. X-ray and NMR structure of human Bcl-xL, an inhibitor of programmed cell death. Nature. May 23, 1996;381(6580):335-41.
Muir, Semisynthesis of proteins by expressed protein ligation. Annu Rev Biochem. 2003;72:249-89. Epub Feb. 27, 2003.
Muller, P. Glossary of terms used in physical organic chemistry. Pure and Applied Chemistry, 1994, vol. 66, pp. 1077-1184.
Mulqueen et al. Synthesis of the thiazoline-based siderophore (5)-desferrithiocin. 1993;48(24):5359-5364.
Muppidi et al., Conjugation of spermine enhances cellular uptake of the stapled peptide-based inhibitors of p53-Mdm2 interaction. Bioorg Med Chem Lett. Dec. 15, 2011;21(24):7412-5. doi: 10.1016/j.bmc1.2011.10.009. Epub Oct. 12, 2011.
Mustapa, et al. Synthesis of a Cyclic Peptide Containing Norlanthionine: Effect of the Thioether Bridge on Peptide Conformation. J. Org. Chem. 2003;68(21):8193-8198.
Nair et al., X-ray structures of Myc-Max and Mad-Max recognizing DNA. Molecular bases of regulation by proto-oncogenic transcription factors. Cell. Jan. 24, 2003;112(2):193-205.
Nam et al., Structural basis for cooperativity in recruitment of MAML coactivators to Notch transcription complexes. Cell. Mar. 10, 2006;124(5):973-83.
Nelson & Kallenbach, "Persistence of the α-Helix Stop Signal in the S-Peptide in Trifluoroethanol Solutions," Biochemistry 28:5256-5261 (1989).
Ngo et al. Computational complexity, protein structure prediction and the Levinthal Paradox.ln: The Protein Folding Problem and Tertiary Structure Prediction. K.Merz, Jr. and S. LeGrand, eds., 1994, pp. 491-495.
Noah, et al. A cell-based luminescence assay is effective for high-throughput screening of potential influenza antivirals. Antiviral Res. Jan. 2007;73(1):50-9. Epub Jul. 28, 2006.
Notice of allowance dated Aug. 6, 2012 for U.S. App. No. 12/796,212.
Notice of allowance dated Dec. 12, 2017 for U.S. Appl. No. 15/287,513.
Notice of allowance dated Feb. 15, 2017 for U.S. Appl. No. 14/852,368.
Notice of allowance dated Jan. 27, 2014 for U.S. Appl. No. 12/233,555.
Notice of allowance dated Jan. 7, 2015 for U.S. Appl. No. 13/370,057.
Notice of allowance dated Jul. 19, 2016 for U.S. Appl. No. 14/068,844.
Notice of allowance dated Jul. 21, 2016 for U.S. Appl. No. 14/677,679.
Notice of Allowance dated Jul. 22, 2015 for U.S. Appl. No. 14/070,367.
Notice of allowance dated Jul. 28, 2014 for U.S. Appl. No. 13/680,905.
Notice of allowance dated Jul. 7, 2009 for U.S. Appl. No. 10/981,873.
Notice of allowance dated Jun. 1, 2016 for U.S. Appl. No. 14/070,354.
Notice of allowance dated Mar. 2, 2017 for U.S. Appl. No. 14/852,368.
Notice of allowance dated Mar. 22, 2010 for U.S. Appl. No. 11/148,976.
Notice of allowance dated Mar. 29, 2017 for U.S. Appl. No. 14/852,368.
Notice of allowance dated May 18, 2016 for U.S. Appl. No. 14/070,354.
Notice of allowance dated May 4, 2004 for U.S. Appl. No. 09/574,086.
Notice of allowance dated May 8, 2012 for U.S. Appl. No. 12/182,673.
Notice of allowance dated Oct. 23, 2015 for U.S. Appl. No. 13/252,751.
Obayashi, E., et al., The structural basis for an essential subunit interaction in influenza virus RNA polymerase. Nature, 2008. 454: p. 1127-31.
Obayashi, et al. The structural basis for an essential subunit interaction in influenza virus RNA polymerse. Nature. Aug. 28, 2008;454(7208):1127-31. Epub Jul. 27, 2008.
Office action dated Apr. 17, 2017 for U.S. Appl. No. 15/287,513.
Office action dated Apr. 18, 2011 for U.S. Appl. No. 12/182,673.
Office action dated Apr. 26, 2012 for U.S. Appl. No. 13/097,930.
Office action dated Apr. 28, 2016 for U.S. Appl. No. 14/677,679.
Office action dated Aug. 10, 2009 for U.S. Appl. No. 11/957,325.
Office action dated Aug. 11, 2009 for U.S. Appl. No. 12/140,241.
Office action dated Aug. 19, 2010 for U.S. Appl. No. 12/037,041.
Office action dated Aug. 30, 2017 for U.S. Appl. No. 15/287,513.
Office action dated Aug. 9, 2010 for U.S. Appl. No. 12/182,673.
Office action dated Dec. 13, 2012 for U.S. Appl. No. 12/690,076.
Office action dated Dec. 19, 2014 for U.S. Appl. No. 14/068,844.
Office action dated Dec. 29, 2011 for U.S. Appl. No. 12/233,555.
Office action dated Dec. 5, 2008 for U.S. Appl. No. 10/981,873.
Office action dated Dec. 7, 2015 for U.S. Appl. No. 14/677,679.
Office action dated Feb. 17, 2011 for U.S. Appl. No. 12/796,212.
Office action dated Feb. 24, 2015 for U.S. Appl. No. 13/252,751.
Office action dated Feb. 4, 2014 for U.S. Appl. No. 13/370,057.
Office action dated Feb. 5, 2016 for U.S. Appl. No. 14/068,844.
Office action dated Feb. 6, 2014 for U.S. Appl. No. 13/680,905.
Office action dated Feb. 9, 2012 for U.S. Appl. No. 12/420,816.
Office action dated Jan. 10, 2013 for U.S. Appl. No. 13/120,370.
Office action dated Jan. 26, 2009 for U.S. Appl. No. 11/148,976.
Office action dated Jan. 27, 2014 for U.S. Appl. No. 13/129,118.
Office Action dated Jan. 30, 2008 for U.S. Appl. No. 10/981,873.
Office action dated Jul. 15, 2013 for U.S. Appl. No. 13/570,146.
Office action dated Jul. 17, 2017 for U.S. Appl. No. 14/853,894.
Office action dated Jul. 21, 2014 for U.S. Appl. No. 13/370,057.
Office action dated Jul. 24, 2015 for U.S. Appl. No. 13/252,751.
Office action dated Jul. 25, 2017 for U.S. Appl. No. 15/200,422.
Office action dated Jul. 30, 2013 for U.S. Appl. No. 13/097,930.
Office action dated Jun. 14, 2013 for U.S. Appl. No. 12/478,504.
Office action dated Jun. 18, 2015 for U.S. Appl. No. 14/068,844.
Office action dated Jun. 28, 2012 for U.S. Appl. No. 12/233,555.
Office action dated Jun. 28, 2013 for U.S. Appl. No. 13/370,057.
Office action dated Mar. 18, 2009 for U.S. Appl. No. 11/678,836.
Office action dated Mar. 18, 2013 for U.S. Appl. No. 13/097,930.
Office action dated Mar. 18, 2015 for U.S. Appl. No. 14/070,367.
Office action dated Mar. 22, 2013 for U.S. Appl. No. 12/233,555.
Office action dated Mar. 22, 2013 for U.S. Appl. No. 13/120,376.
Office action dated Mar. 26, 2015 for U.S. Appl. No. 14/070,354.
Office action dated May 10, 2010 for U.S. Appl. No. 11/957,325.
Office action dated May 19, 2010 for U.S. Appl. No. 12/140,241.
Office action dated May 26, 2017 for U.S. Appl. No. 14/853,894.
Office action dated Nov. 15, 2017 for U.S. Appl. No. 15/240,505.
Office action dated Nov. 16, 2015 for U.S. Appl. No. 14/070,354.
Office action dated Nov. 25, 2009 for U.S. Appl. No. 11/148,976.
Office action dated Nov. 5, 2002 for U.S. Appl. No. 09/574,086.
Office action dated Nov. 8, 2012 for U.S. Appl. No. 13/120,386.
Office action dated Oct. 15, 2012 for U.S. Appl. No. 13/097,930.
Office action dated Oct. 18, 2011 for U.S. Appl. No. 12/796,212.
Office action dated Oct. 24, 2016 for U.S. Appl. No. 14/718,288.
Office action dated Oct. 31, 2014 for U.S. Appl. No. 13/370,057.
Office action dated Sep. 20, 2016 for U.S. Appl. No. 14/852,368.
Office action dated Sep. 20, 2016 for U.S. Appl. No. 14/853,894.
Office action dated Sep. 23, 2013 for U.S. Appl. No. 13/680,905.
Office action dated Sep. 5, 2017 for U.S. Appl. No. 15/093,869.
Office action dated Sep. 7, 2017 for U.S. Appl. No. 15/093,426.
O'Neil & DeGrado, "A Thermodynamic Scale for the Helix-forming Tendencies of the Commonly Occurring Amino Acids," Science 250:646-651(1990).
Or et al. Cysteine alkylation in unprotected peptides: synthesis of a carbavasopressin analogue by intramolecular cystein alkylation. J. Org. Chem. Apr. 1991;56(9):3146-3149.
Ösapay & Taylor, "Multicyclic Polypeptide Model Compounds. 2. Synthesis and Conformational Properties of a Highly α-Helical Uncosapeptide Constrained by Three Side-chain to Side-chain Lactam Bridges," J. Am. Chem. Soc. 114:6966-6973 (1992).
O'Shea et al., "Mechanism of Specificity in the Fos-Jun Oncoprotein Heterodimer," Cell 68:699-708 (1992).
Ou, et al. Review of the role of D14-Notch signaling parthway in angiogenesis. Basic Medical Sciences and Clinics. 2008; 28(1):98-105 (in Chinese with English abstract).
Pangborn et al., "Safe and Convenient Procedure for Solvent Purification," Organometallics 15:1518-1520 (1996).
Paquette, L.A., ed. Encyclopedia of Reagents for Organic Synthesis. New York; John Wiley & Sons; 1995.
Patgiri et al. An orthosteric inhibitor of the Ras-Sos interaction. Nat Chem Bio 7:585-587 (2011).
Patgiri, et al. A hydrogen bond surrogate approach for stabilization of short peptide sequences in alpha-helical conformation. Acc Chem Res. Oct. 2008;41(10):1289-300. Epub Jul. 17, 2008.
Patgiri, et al. Solid phase synthesis of hydrogen bond surrogate derived alpha-helices: resolving the case of a difficult amide coupling. Org Biomol Chem. Apr. 21, 2010;8(8):1773-6.
Pattenden, et al. Enantioselective synthesis of 2-alkyl substituted cysteines. 1993;49(10):2131-2138.
Pattenden, et al. Naturally occurring linear fused thiazoline-thiazole containing metabolites: total synthesis of (−)-didehydromirabazole A, a cytotoxic alkaloid from blue-green algae. J Chem Soc. 1993;14:1629-1636.
Pellois et al., Semisynthetic proteins in mechanistic studies: using chemistry to go where nature can't. Curr Opin Chem Biol. Oct. 2006;10(5):487-91. Epub Aug. 28, 2006.
Peryshkov, et al. Z-Selective olefin metathesis reactions promoted by tungsten oxo alkylidene complexes. J Am Chem Soc. Dec. 28, 2011;133(51):20754-7. doi: 10.1021/ja210349m. Epub Nov. 30, 2011.
Petros et al., "Rationale for Bcl-xL/Bad Peptide Complex Formation from Structure, Mutagenesis, and Biophysical Studies," Protein Sci. 9:2528-2534 (2000).
Phelan, et al. A General Method for Constraining Short Peptides to an α-Helical Conformation. J. Am. Chem. Soc. 1997;119:455-460.
Punna, et al. Head-to-tail peptide cyclodimerization by copper-catalyzed azide-alkyne cycloaddition. Angew Chem Int Ed Engl. Apr. 8, 2005;44(15):2215-20.
Qian & Schellman, "Helix-coil Theories: A Comparative Study for Finite Length Polypeptides," J. Phys. Chem. 96:3987-3994 (1992).
Qiu et al., Convenient, Large-Scale Asymmetric Synthesis of Enantiomerically Pure trans-Cinnamylglycine and -a-Alanine. Tetrahedron. 2000;56:2577-82.
Rankin, et al. The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ. Apr. 2008;15(4):678-85.
Rasmussen, et al. Ruthenium-catalyzed cycloaddition of aryl azides and alkynes. Org Lett. Dec. 20, 2007;9(26):5337-9.
Rich et al., Synthesis of the cytostatic cyclic tetrapeptide, chlamydocin. Tetranderon Letts. 1983;24(48):5305-08.
Rink, et al. Lantibiotic Structures as Guidelines for the Design of Peptides That Can Be Modified by Lantibiotic Enzymes. Biochemistry. 2005; 44:8873-8882.
Robert, A hierarchical "nesting" approach to describe the stability of alpha helices with side-chain interactions. Biopolymers. 1990;30(3-4):335-47.
Roberts, et al. Efficient synthesis of thioether-based cyclic peptide libraries. Tetrahedon Letters. 1998; 39: 8357-8360.
Roberts, et al. Examination of methodology for the synthesis of cyclic thioether peptide libraries derived from linear tripeptides. J Pept Sci. Dec. 2007;13(12):811-21.
Roehrl et al., "A General Framework for Development and Data Analysis of Competitive High-throughput Screens for Small-molecule Inhibitors of Protein-Protein Interactions by Fluorescence Polarization," Biochemistry 43:16056-16066 (2004).
Roehrl et al., "Discovery of Small-molecule Inhibitors of the NFAT-Calcineurin Interaction by Competitive High-throughput Fluorescence Polarization Screening," Biochemistry 43:16067-16075 (2004).
Roice, et al. High Capacity Poly(ethylene glycol) Based Amino Polymers for Peptide and Organic Synthesis. QSAR & Combinatorial Science. 2004;23(8):662-673.
Rojo, et al. Macrocyclic peptidomimetic inhibitors of β-secretase (BACE): First X-ray structure of a macrocyclic peptidomimetic-BACE complex. Bioorg. Med. Chem. Lett. 2006; 16:191-195.
Roof, et al. Mechanism of action and structural requirements of constrained peptide inhibitors of RGS proteins. Chem Biol Drug Des. Apr. 2006;67(4):266-74.
Rostovtsev et al. A stepwise huisgen cycloaddition process: copper (i)-catalyzed regioselective "ligation" of azides and terminal alkynes. Angew. Chem. Int. Ed. Engl. 41(14):2596-2599 (2002).
Rostovtsev, et al. A stepwise huisgen cycloaddition process: copper(I)-catalyzed regioselective "ligation" of azides and terminal alkynes. Angew Chem Int Ed Engl. Jul. 15, 2002;41(14):2596-9.
Ruan et al., "Metal Ion Enhanced Helicity in Synthetic Peptides Containing Unnatural, Metal-ligating Residues," J. Am. Chem. Soc. 112:9403-9404 (1990).
Rudinger J, "Characteristics of the amino acids as components of a peptide hormone sequence," Peptide Hormones, JA Parsons Edition, University Park Press, Jun. 1976, pp. 1-7.
Ruffolo and Shore. Bcl-2 Selectively Interacts with the BID-Induced Open Conformer of BAK, Inhibiting BAK Auto-Oligomerization. J. Biol. Chern. 2003;278(27):25039-25045.
Rushe, et al. Structure of a NEMO/IKK-associating domain reveals architecture of the interaction site. Structure. May 2008;16(5):798-808.
Rutledge et al., "A View to a Kill: Ligands for Bcl-2 Family Proteins," Curr. Opin. Chem. Biol. 6:479-485 (2002).
Sali et al., Stabilization of protein structure by interaction of alpha-helix dipole with a charged side chain. Nature. Oct. 20, 1988;335(6192):740-3.
Sanchez-Garcia, et al. Tumorigenic activity of the BCR-ABL oncogenes is mediated by BCL2. Proc Natl Acad Sci U S A. Jun. 6, 1995;92(12):5287-91.
Sattler et al. Structure of Bcl-xL-Back peptide complex: recognition between regulators of apoptosis. Science. 275:983-986 (1997).
Schafmeister et al. An all-hydrocarbon crosslinking system for enhancing the helicity and metabolic stability of peptides. J. Am Chem. Soc. 2000;122:5891-5892.
Scheffzek et al. The Ras-RasGAP complex: structural basis for GTPase activation and its loss in oncogenic Ras mutants. Science 277(5324):333-338 (1997).
Schinzel et al., The phosphate recognition site of Escherichia coli maltodextrin phosphorylase. FEBS Lett. Jul. 29, 1991;286(1-2):125-8.
Schmiedeberg et al. Reversible backbone protection enables combinatorial solid-phase ring-closing metathesis reaction (RCM) in peptides. Org Lett. Jan. 10, 2002;4(1):59-62.
Scholtz et al., The mechanism of alpha-helix formation by peptides. Annu Rev Biophys Biomol Struct. 1992;21:95-118.
Scorrano, et al. A distinct pathway remodels mitochondrial cristae and mobilizes cytochrome c during apoptosis. Dev Cell. Jan. 2002;2(1):55-67.
Scott et al., Evidence of insulin-stimulated phosphorylation and activation of the mammalian target of rapamycin mediated by a protein kinase B signaling pathway. Proc Natl Acad Sci U S A. Jun. 23, 1998;95(13):7772-7.
Scott, et al. A Solid-Phase Synthetic Route to Unnatural Amino Acids with Diverse Side-Chain Substitutions. Journal of Organic Chemistry. 2002, vol. 67, No. 9, pp. 2960-2969.
Seebach, et al. Beta-peptidic peptidomimetics. Acc Chem Res. Oct. 2008;41(10):1366-75. doi: 10.1021/ar700263g. Epub Jun. 26, 2008.
Seebach, et al. Self-Regeneration of Stereocenters (SRS)-Applications, Limitations, and Abandonment of a Synthetic Principle. Angewandte Chemie International Edition in English. 1996;35(23-24):2708-2748.
Seebach, et al. Self-Regeneration of Stereocenters (SRS)—Applications, Limitations, and Abandonment of a Synthetic Principle. Angewandte Chemie International Edition in English. 1996;35(23-24):2708-2748.
Seebeck, et al. Ribosomal synthesis of dehydroalanine-containing peptides. J Am Chem Soc. Jun. 7, 2006;128(22):7150-1.
Shepherd et al., "Single Turn Peptide Alpha Helices with Exceptional Stability in Water," J. Am. Chem. Soc. 127:2974-2983 (2005).
Shi, et al. The role of arsenic-thiol interactions in metalloregulation of the ars operant. J Biol Chem. Apr. 19, 1996;271(16):9291-7.
Sia et al., "Short Constrained Peptides that Inhibit HIV-1 Entry," Proc. Nat'l Acad. Sci. USA 99(23):14664-14669 (2002).
Singh, et al. Efficient asymmetric synthesis of (S)- and (R)-N-Fmoc-S-trityl-alpha-methylcysteine using camphorsultam as a chiral auxiliary . . . J Org Chem. Jun. 25, 2004;69(13):4551-4.
Smith, et al. Design, Synthesis, and Binding Affinities of Pyrrolinone-Based Somatostatin Mimetics. Organic Letters. Jan. 8, 2005, vol. 7, No. 3, pp. 399-402, plus Supporting Information, pp. S1-S39.
Solution phase synthesis from http://www.combichemistry.com/solution_phase_synthesis.html. p. 1. Accessed Aug. 6, 2009.
Sparey et al., Cyclic sulfamide gamma-secretase inhibitors. Bioorg Med Chem Lett. Oct. 1, 2005;15(19):4212-6.
Spierings, et al. Connected to death: the (unexpurgated) mitochondrial pathway of apoptosis. Science. 2005; 310:66-67.
Stewart, et al. Cell-penetrating peptides as delivery vehicles for biology and medicine. Org Biomol Chem. Jul. 7, 2008;6(13):2242-55. doi: 10.1039/b719950c. Epub Apr. 15, 2008.
Still et al., "Rapid Chromatographic Technique for Preparative Separations with Moderate Resolution," J. Org. Chem. 43(14):2923-2925 (1978).
Strickland, et al. Use of cell permeable NBD peptides for suppression of inflammation. Ann Rheum Dis. Nov. 2006;65 Suppl 3:iii75-82.
Strnad, et al. IkappaB kinase inhibitors for treating autoimmune and inflammatory disorders: potential and challenges. Trends Pharmacol Sci. Mar. 2007;28(3):142-8. Epub Feb. 6, 2007.
Sugiyama K, et al. Structural insight into the essential PB1-PB2 subunit contact of the influenza virus RNA polymerase. The EMBO Journal. 2009;28(12):1803-1811. doi:10.1038/emboj.2009.138.
Sugiyama K, et al. Structural insight into the essential PB1—PB2 subunit contact of the influenza virus RNA polymerase. The EMBO Journal. 2009;28(12):1803-1811. doi:10.1038/emboj.2009.138.
Sugiyama, et al. Structural insight into the essential PB1-PB2 subunit contact of the influenza virus RNA polymerase. EMBO J. Jun. 17, 2009;28(12):1803-11. Epub May 21, 2009.
Suzuki, et al. Structure of Bax: coregulation of dimer formation and intracellular localization. Cell. Nov. 10, 2000;103(4):645-54.
Szewczuk, et al. Synthesis and Biological activity of new conformationally restricted analogues of pepstatin. Int. J. Peptide Protein. Res. 1992; 40:233-242.
Tam, et al. Protein prosthesis: 1,5-disubstituted[1,2,3]triazoles as cis-peptide bond surrogates. J Am Chem Soc. Oct. 24, 2007;129(42):12670-1.
Tanaka, Design and synthesis of non-proteinogenic amino acids and secondary structures of their peptides. Yakugaku Zasshi. Oct. 2006:126(10):931-44. Japanese.
Taylor. The synthesis and study of side-chain lactam-bridged peptides. Biopolymers. 2002;66(1):49-75.
Titus, et al. Human K/natural killer cells targeted with hetero-cross-linked antibodies specifically lyse tumor cells in vitro and prevent tumor growth in vivo. J Immunol. Nov. 1, 1987;139(9):3153-8.
Tolbert et al., New methods for proteomic research: preparation of proteins with N-terminal cysteines for labeling and conjugation. Angew Chem Int Ed Engl. Jun. 17, 2002;41(12):2171-4.
Toniolo, Conformationally restricted peptides through short-range cyclizations. Int J Pept Protein Res. Apr. 1990;35(4):287-300.
Tornoe et al., Peptidotriazoles on solid phase: [1,2,3]-triazoles by regiospecific copper(i)-catalyzed 1,3-dipolar cycloadditions of terminal alkynes to azides. J Org Chem. May 3, 2002;67(9):3057-64.
Torres, et al. Peptide tertiary structure nucleation by side-chain crosslinking with metal complexation and double "click" cycloaddition. Chembiochem. Jul. 21, 2008;9(11):1701-5.
Trnka & Grubbs, "The Development of L2X2Ru=CHR Olefin Metathesis Catalysts: An Organometallic Success Story," Acc. Chem. Res. 34:18-29 (2001).
Tsuruzoe et al., Insulin receptor substrate 3 (IRS-3) and IRS-4 impair IRS-1- and IRS-2-mediated signaling. Mol Cell Biol. Jan. 2001;21(1):26-38.
Tugyi, et al. The effect of cyclization on the enzymatic degradation of herpes simplex virus glycoprotein D derived epitope peptide. J Pept Sci. Oct. 2005;11(10):642-9.
Turner et al., "Mitsunobu Glycosylation of Nitrobenzenesulfonamides: Novel Route to Amadori Rearrangement Products," Tetrahedron Lett. 40:7039-7042 (1999).
Tyndall et al. Macrocycles mimic the extended peptide conformation recognized by aspartic, serine, cysteine and metallo proteases. Curr Med Chem. Jul. 2001;8(8):893-907.
Tyndall et al., "Proteases Universally Recognize Beta Strands in Their Active Sites," Chem. Rev. 105:973-999 (2005).
U.S. Appl. No. 15/093,426 Notice of Allowance dated Feb. 27, 2018.
U.S. Appl. No. 15/093,426 Office action dated Jan. 8, 2018.
U.S. Appl. No. 15/093,869 Office action dated Jan. 22, 2018.
Ueki et al., Increased insulin sensitivity in mice lacking p85beta subunit of phosphoinositide 3-kinase. Proc Natl Acad Sci U S A. Jan. 8, 2002;99(1):419-24. Epub Dec. 18, 2001.
Ueki et al., Positive and negative regulation of phosphoinositide 3-kinase-dependent signaling pathways by three different gene products of the p85alpha regulatory subunit. Mol Cell Biol. Nov. 2000;20(21):8035-46.
Vaickus et al., Immune markers in hematologic malignancies. Crit Rev Oncol Hematol. Dec. 1991; 11(4):267-97.
Van Maarseveen, et al. Efficient route to C2 symmetric heterocyclic backbone modified cyclic peptides. Org Lett. Sep. 29, 2005;7(20):4503-6.
Vassilev, et al. In Vivo Activation of the p53 Pathway by Small-molecule Antagonists of MDM2. Science. 2004; 303:844-848.
Verdine et al. The challenge of drugging undruggable targets in cancer: lessons learned from targeting BCL-2 family members. Clin Cancer Res. 13(24):7264-7270 (2007).
Verdine et al., Stapled peptides for intracellular drug targets. Methods Enzymol. 2012;503:3-33. doi: 10.1016/B978-0-12-396962-0.00001-X.
Viallet, et al. Tallimustine is inactive in patients with previously treated small cell lung cancer. A phase II trial of the National Cancer Institute of Canada Clinical Trials Group. Lung Cancer. Nov. 1996;15(3):367-73.
Vila-Perello, et al. A minimalist design approach to antimicrobial agents based on a thionin template. J Med Chem. Jan. 26, 2006;49(2):448-51.
Voet D, Voet JG, Biochemistry, Second Edition, John Wiley & Sons, Inc., 1995, pp. 235-241.
von Itzstein et al, "Rational design of potent sialidase-based inhibitors of influenza virus replication," Nature, Jun. 3, 1993, 363: 418-423.
Walensky et al., A stapled BID BH3 helix directly binds and activates BAX. (2006) Mol Cell 24:199-210.
Walensky, et al. Activation of apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science. Sep. 3, 2004;305(5689):1466-70.
Walensky, et al. Hydrocarbon-stapled peptides: principles, practice, and progress. J Med Chem. Aug. 14, 2014;57(15):6275-88. doi: 10.1021/jm4011675. Epub Mar. 6, 2014.
Walker, et al. General method for the synthesis of cyclic peptidomimetic compounds. Tetrahedron Letters. 2001; 42(34):5801-5804.
Wang et al. Cell permeable Bcl-2 binding peptides: a chemical approach to apoptosis induction in tumor cells. Cancer Res. Mar. 15, 2000;60(6):1498-502.
Wang et al. Enhanced metabolic stability and protein-binding properties of artificial alpha helices derived from a hydrogen-bond surrogate: application to Bcl-xL. Angew Chem Int Ed Engl. Oct. 14, 2005;44(40):6525-9.
Wang et al., "Recognition of a Protein Receptor with the Hydrogen Bond Surrogate-based Artificial Alpha-helices," American Chemical Society Meeting, San Diego (Mar. 2005) (poster).
Wang et al., "Recognition of a Protein Receptor with the Hydrogen Bond Surrogate-based Artificial Alpha-helices," Chemical Biology Symposium, Hunter College (Jan. 2005) (poster).
Wang, et al. "Click" synthesis of small molecule probes for activity-based fingerprinting of matrix metalloproteases. Chem Commun (Camb). Sep. 28, 2006;(36):3783-5.
Wang, et al. BID: a novel BH3 domain-only death agonist. Genes Dev. Nov. 15, 1996;10(22):2859-69.
Wang, et al. Evaluation of biologically relevant short alpha-helices stabilized by a main-chain hydrogen-bond surrogate. J Am Chem Soc. Jul. 19, 2006;128(28):9248-56.
Wang, et al. Nucleation and stability of hydrogen-bond surrogate-based alpha-helices. Org Biomol Chem. Nov. 21, 2006;4(22):4074-81.
Wei, et al. tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c. Genes Dev. Aug. 15, 2000;14(16):2060-71.
Wels, et al. Synthesis of a novel potent cyclic peptide MC4-ligand by ring-closing metathesis. Bioorg. Med. Chem. Lett. 2005; 13: 4221-4227.
Weng et al., Growth suppression of pre-T acute lymphoblastic leukemia cells by inhibition of notch signaling. Mol Cell Biol. Jan. 2003;23(2):655-64.
Wild et al., "Peptides Corresponding to a Predictive α-Helical Domain of Human Immunodeficiency Virus Type 1 gp4I are Potent Inhibitors of Virus Infection," Proc. Nat'l Acad. Sci. USA 91:9770-9774 (1994).
Williams and Im. Asymmetric Synthesis of Nonsubstituted and α,α-Disubstituted α-Amino Acids via Disatereoselective Glycine Enolate Alkylations. JACS. 1991;113:9276-9286.
Williams, et al. A novel family of cyclic peptide antagonists suggests that N-cadherin specificity is determined by amino acids that flank the HAV motif. J Biol Chem. Feb. 11, 2000;275(6):4007-12.
Williams, et al. Dimeric versions of two short N-cadherin binding motifs (HAVDI and INPISG) function as N-cadherin agonists. J Biol Chem. Feb. 8, 2002;277(6):4361-7.
Woodfin, et al. Interaction of the amino-terminus of an influenza virus protein with mitochondria. Arch Biochem Biophys. Nov. 1, 1993;306(2):427-30.
Wu, et al. Modulation of Notch signaling by mastermind-like (MAML) transcriptional co-activators and their involvement in tumorigenesis. Semin Cancer Biol. Oct. 2004;14(5):348-56.
Wu, et al. Regiospecific Synthesis of 1,4,5-Trisubstituted-1,2,3-triazole via One-Pot Reaction Promoted by Copper(I) Salt. Synthesis. 2005(8): 1314-1318.
Wu, et al. Studies on New Strategies for the Synthesis of Oligomeric 1,2,3-Triazoles. Synlett 2006(4): 0645-0647.
Wunderlich, et al. Identification of a PA-binding peptide with inhibitory activity against influenza A and B virus replication. PLoS One. Oct. 20, 2009;4(10):e7517. doi: 10.1371/journal.pone.0007517.
Yang et al. Synthesis and helical structure of lactam bridged BH3 peptides derived from pro-apoptotic Bcl-2 family proteins. Bioorg Med Chem Lett. Mar. 22, 2004;14(6):1403-6.
Yang, et al. Calculation of protein conformation from circular dichroism. Methods Enzymol. 1986;130:208-69.
Yang, et al. Synthesis and helical structure of lactam bridged BH3 peptides derived from pro-apoptotic Bcl-2 family proteins. Bioorganic & medicinal chemistry letters. 2004;14(6):1403-1406.
Yin et al., "Terphenyl-based Helical Mimetics That Disrupt the p53/HDM2 Interaction," Angew. Chem. Int. Ed. 44:2704-2707 (2005).
Yu, et al. Synthesis of macrocyclic natural products by catalyst-controlled stereoselective ring-closing metathesis. Nature. Nov. 2, 2011;479(7371):88-93. doi: 10.1038/nature10563.
Zamzami et al. The thiol crosslinking agent diamide overcomes the apoptosis-inhibitory effect of Bcl-2 by enforcing mitochondrial permeability transition. Oncogene. Feb. 26, 1998;16(8):1055-63.
Zhang, et al. 310 Helix versus alpha-helix: a molecular dynamics study of conformational preferences of Aib and Alanine. J. American Cancer Society. Dec. 1994; 116(26):11915-11921.
Zhang, et al. Development of a High-throughput Fluorescence Polarization Assay for Bcl-xL. Anal. Biochem. 2002; 307:70-75.
Zhang, et al. Ruthenium-catalyzed cycloaddition of alkynes and organic azides. J Am Chem Soc. Nov. 23, 2005;127(46):15998-9.
Zimm & Bragg, "Theory of the Phase Transition between Helix and Random Coil in Polypeptide Chains," J. Chem. Phys. 31(2):526-535 (1959).
Zitzow, et al. Pathogenesis of avian influenza A (H5N1) viruses in ferrets. J Virol. May 2002;76(9):4420-9.

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10328117B2 (en) 2006-12-14 2019-06-25 Aileron Therapeutics, Inc. Bis-sulfhydryl macrocyclization systems
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
US10300109B2 (en) 2009-09-22 2019-05-28 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10703780B2 (en) 2010-08-13 2020-07-07 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10308699B2 (en) 2011-10-18 2019-06-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10213477B2 (en) 2012-02-15 2019-02-26 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10669230B2 (en) 2012-11-01 2020-06-02 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US10246491B2 (en) 2013-03-06 2019-04-02 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and use thereof in regulating HIF1alpha
US10471120B2 (en) 2014-09-24 2019-11-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10905739B2 (en) 2014-09-24 2021-02-02 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
US12059413B2 (en) 2016-11-02 2024-08-13 The Research Foundation For The State University Of New York Methods of inhibiting viruses using compositions targeting TSG101-ubiquitin interaction
US11091522B2 (en) 2018-07-23 2021-08-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof

Also Published As

Publication number Publication date
US20120040889A1 (en) 2012-02-16
EP2376100A4 (en) 2013-08-14
US20160250278A1 (en) 2016-09-01
JP2012515172A (en) 2012-07-05
AU2010204648B2 (en) 2016-09-01
CA2744088A1 (en) 2010-07-22
CN102256615A (en) 2011-11-23
EP2376100B1 (en) 2017-10-25
US20160068573A1 (en) 2016-03-10
US20180339014A1 (en) 2018-11-29
AU2010204648A1 (en) 2010-07-22
WO2010083347A2 (en) 2010-07-22
BRPI1006139A2 (en) 2017-05-30
US9175047B2 (en) 2015-11-03
US20160251399A1 (en) 2016-09-01
EP2376100A2 (en) 2011-10-19
WO2010083347A3 (en) 2010-09-10

Similar Documents

Publication Publication Date Title
US10022422B2 (en) Peptidomimetic macrocycles
US10059741B2 (en) Peptidomimetic macrocycles
US20210299211A1 (en) Peptidomimetic macrocycles and formulations thereof
WO2017205786A1 (en) Cell permeable peptidomimetic macrocycles
US9290545B2 (en) Compositions and methods for the treatment of viral infections
WO2022202816A1 (en) Peptide and peptide-containing composition
JP2022088370A (en) Peptide as antiviral agent, and uses therefor
US11376306B2 (en) Peptides and uses therefor as antiviral agents
US9464113B2 (en) Anti-heparan sulfate peptides that block herpes simplex virus infection in vivo
WO2020171028A1 (en) Hemagglutinin-binding peptide
Zima Preparation of Influenza Neuraminidase and Polymerase Inhibitors
CN117062826A (en) Antiinfective bicyclic peptide ligands
JP2024533273A (en) Antiviral structurally stapled sars-cov-2 peptide-cholesterol conjugates and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: AILERON THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NASH, HUW M.;ANNIS, DAVID ALLEN;SIGNING DATES FROM 20160808 TO 20160912;REEL/FRAME:040594/0884

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20220717